Abstract
Drugs of abuse such as morphine or marijuana exert their effects through the activation of G-protein-coupled receptors (GPCRs), the opioid and cannabinoid receptors, respectively. Moreover, interactions between either of these receptors have been shown to be involved in the rewarding effects of drugs of abuse. Recent advances in the field, using a variety of approaches, have demonstrated that many GPCRs, including opioid, cannabinoid, and dopamine receptors, can form associations between different receptor subtypes or with other GPCRs to form heteromeric complexes. The formation of these complexes, in turn, leads to the modulation of the properties of individual protomers. The development of tools that can selectively disrupt GPCR heteromers as well as monoclonal antibodies that can selectively block signaling by specific heteromer pairs has indicated that heteromers involving opioid, cannabinoid, or dopamine receptors may play a role in various disease states. In this review, we describe evidence for opioid, cannabinoid, and dopamine receptor heteromerization and the potential role of GPCR heteromers in pathophysiological conditions.
1. INTRODUCTION
Morphine, an alkaloid extracted from the poppy Papaver somniferum, and Δ9-tetrahydrocannabinol (also known as THC or more commonly as marijuana), the main psychoactive extract from Cannabis sativum, are commonly abused drugs due to their mood-altering effects. Morphine is the most commonly used drug in the treatment of chronic pain, while THC is used in the alleviation of neuropathic pain and for the reduction of eye pressure in glaucoma patients.1–5 However, long-term use of these drugs leads to a number of undesirable side effects, including the development of tolerance and addiction. In the latter context, several lines of evidence demonstrate the involvement of the dopaminergic system in the rewarding effects of drugs of abuse (reviewed in Ref. 6). Both morphine and THC exert their physiological effects via activation of Gαi/o-coupled receptors, the opioid and cannabinoid receptors, respectively.1,3–5 In an effort to develop drugs that _target these receptors and exert the same physiological effects as morphine or THC but without the undesirable side effects associated with chronic drug use, several groups have investigated whether protein–protein interactions, including receptor heteromerization, could modulate the function of individual receptors. Here, we describe heteromerization as associations between different subtypes of a receptor or between two different receptors. Although heteromers involving opioid, cannabinoid, or dopamine receptors have been described, not much is known about their physiological role. In this context, studies using peptides that disrupt specific heteromers, or antibodies that selectively block heteromer signaling, suggest that GPCR heteromers may play a role in a number of pathologies.7–11 While additional GPCR heteromers associated with other drugs of abuse have been reported, for the sake of brevity and conciseness, in this review we have focused only on heteromers that contain either opioid or cannabinoid receptors as one of the partners. We have also included dopamine receptors since they are the primary effectors of the action of stimulants such as cocaine and amphetamines. These are described below along with the putative role of these heteromers in the development of tolerance and other disease states.
1.1. Opioid receptors
Opioid receptors are members of the rhodopsin family of GPCRs and their signal transduction involves activation of Gαi/o proteins, downstream inhibition of adenylyl cyclase activity, the opening of inwardly rectifying K+ channels, and inhibition of Ca2+ channels.1 Molecular cloning studies have identified three types of opioid receptors μ-, δ-, and κ-opioid receptors (reviewed in Ref. 2).
Opioid receptors can be activated by endogenous peptides derived from the processing of the large precursor proteins, proopiomelanocortin, proenkephalin, and prodynorphin, by prohormone convertases via the classic peptide-processing pathway12 to generate mainly β-endorphin, Met- and Leu-enkephalin, and dynorphins.13 It is generally thought that enkephalins exert their biological effects primarily by activating δ opioid receptors (DOR), β-endorphin by activating μ opioid receptors (MOR), and dynorphins by activating κ opioid receptors (KOR).13,14 In addition, opioid receptors can also be activated by drugs of abuse such as morphine and heroin.15 Agonist-mediated activation of opioid receptors leads to the modulation of many biological functions including pain perception, motivation, locomotion, hormone secretion, and reward.1,2 Studies using genetic knockdown have further elucidated the role of each opioid receptor type. These studies have implicated MOR in the analgesic effects of morphine and in the rewarding effects resulting from other nonopioid drugs of abuse including cannabinoids,16–20 DOR in the regulation of emotional responses including anxiety and depression,21 and KOR in the dysphoric effects of opioids and cannabinoids as well as in stress-induced relapse.22,23 Together, these studies reveal a broad role for opioid receptors in normal physiology and disease.
1.2. Cannabinoid receptors
Like opioid receptors, cannabinoid receptors are also members of the rhodopsin family of GPCRs and signal via the Gαi/o-signaling pathway, to activate similar downstream signaling cascades.3–5 The two main subtypes of cannabinoid receptors are CB1R and CB2R.3 Recently, the orphan receptor, GPR55, has been proposed as a putative cannabinoid receptor subtype (reviewed in Ref. 24).
CB1R and CB2R are activated by endogenous ligands derived from membrane lipids termed “endocannabinoids” which together with the receptors constitute the “endocannabinoid system” (for review, see Refs. 25–27). The two major lipid-based endocannabinoids are anandamide and 2-arachidonoylglycerol (2-AG)3 that are generated by phospholipases28–32 and degraded by fatty acid amide hydrolase,33,34 cyclooxygenases, lipooxygenases,35,36 and monoacylglycerol lipase.37 Recently, endogenous peptidic agonists of cannabinoid receptors have been identified.38–42 These peptides, RVD-hemopressin and VD-hemopressin, are derived from the α-chain of hemoglobin38 and appear to activate a signal transduction pathway distinct from that of classic CB1R ligands.38 However, very little information is available about the enzymatic steps involved in either the generation or degradation of RVD-hemopressin.
Studies show that the endocannabinoid system plays an important role in many physiological processes including pain perception, learning and memory, appetite control, motor coordination, lipogenesis, and modulation of immune responses (for reviews, see Refs. 25–27,43,44). Studies with knockout mice indicate that the CB1R subtype plays a role in the central effects of cannabinoids, including mood disorders, learning and memory, motor coordination, feeding, control of pain, and stress responses, and in the reinforcing and rewarding effects of drugs of abuse (reviewed in Refs. 45,46). Conversely, the CB2R subtype plays a role in the peripheral effects of cannabinoids, including the development and function of the immune system, embryonic development, bone homeostasis, and pain perception, and plays an antifibrogenic role in liver diseases (reviewed in Ref. 47). These studies imply a broad role for CB1R in central nervous system physiology and CB2R in immune system function.
1.3. Dopamine receptors
Like opioid and cannabinoid receptors, dopamine receptors are members of the family A of GPCRs. Based on their ability to modulate cAMP levels, these receptors are classified as D1- or D2-like receptors (reviewed in Ref. 48). D1-like receptors couple to Gαs/olf proteins and are thought to be exclusively present at the presynapse (reviewed in Ref. 48). Activation of D1-like receptors leads to increases in cAMP levels, activation of cAMP-dependent protein kinases, increase in the phosphorylation levels of DARPP-32 (a molecule involved in the integration of cellular signaling responses to neurotransmitters) and ERK1/2 (the latter particularly in medium spiny neurons, MSNs), increases in L-type Ca2+ channel and NMDA receptor activity, inhibition of N- or P/Q-type Ca2+ channel and K+ channel activity, enhancement of AMPA currents and modulation of GABA currents (reviewed in Refs. 48–50). D2-like receptors couple to Gαi/o proteins and are present both at the pre- and postsynapse (reviewed in Ref. 48). Activation of D2-like receptors decreases cAMP levels and the activity of protein kinase A, decreases the phosphorylation levels of DARPP-32, inhibits ERK1/2 phosphorylation in striatum, decreases the activity of L-, N-, and P/Q-type Ca2+ channels, and activates K+ channels (reviewed in Refs. 48,49). There is some evidence suggesting that, in addition to Gαs/olf and Gαi/o-mediated signaling, dopamine receptors can also signal via Gαq- and G-protein-independent mechanisms (reviewed in Refs. 48–50). However, the mechanisms underlying Gαq-mediated signaling are not completely understood (reviewed in Refs. 48–50). Studies show that dopamine receptors play a role in the modulation of locomotor activity, olfaction, retinal processes, cardiovascular, renal, and immune functions, memory and learning, feeding, sleep regulation, and in reward behaviors, particularly those related to drugs of abuse (reviewed in Ref. 48). Five subtypes of dopamine receptor have been identified. While D1 and D5 dopamine receptors belong to the D1-like receptor type, D2, D3, and D4 receptors belong to the D2-like receptor type (reviewed in Refs. 48–50).
As the name suggests, the endogenous agonist of dopamine receptors is dopamine. Dopamine can be synthesized from phenylalanine and tyrosine by tyrosine hydroxylase (reviewed in Ref. 50). Following synthesis in the cytosol, dopamine is taken into storage vesicles by vesicular monoamine transporters (reviewed in Ref. 50). Dopamine is released at the synapse by Ca2+-mediated fusion of dopamine-containing vesicles with neuronal membranes (reviewed in Ref. 50). The synaptic activity of released dopamine is terminated by its reuptake into presynaptic nerve terminals by dopamine transporters (reviewed in Ref. 50). In the mammalian brain, four major dopaminergic pathways have been identified. These include the nigrostriatal, mesolimbic, mesocortical, and tuberoinfundibular systems (reviewed in Refs. 48–50). Interestingly, it has been shown that MSNs in the striatum and nucleus accumbens can be phenotypically distinguished depending on the expression of D1 or D2 dopamine receptors. D1 dopamine receptors (D1R) are expressed by MSNs that project into the medial globus pallidus and the substantia nigra pars reticulata, D2 dopamine receptors (D2R) are expressed by MSNs that project into the lateral globus pallidus (reviewed in Refs. 48–50), and a small percentage of MSNs coexpress D1R and D2R (reviewed in Refs. 48–50).
In order to elucidate the physiological roles of different dopamine receptor subtypes, knockout mice have been generated. Data with these mice have been the subject of excellent reviews (see Refs. 50–53) and are not described in detail here. Briefly, studies with D1R knockout mice suggest that the latter exhibit alterations in the motivational or attentive aspects of behavior.52 Interestingly, D1R knockout mice exhibit a decrease in the locomotor effects of cocaine.54 Studies with D2R knockout mice suggest that they may represent an animal model for Parkinson’s disease as they recapitulate the extrapyramidal symptoms of the disease.50 Interestingly, while the release of dopamine elicited by drugs of abuse such as morphine and cocaine is higher in D2R knockout mice than in wild-type animals (reviewed in Ref. 55), the rewarding effects of these drugs are attenuated in the knockout mice (reviewed in Ref. 55). D3R knockout mice exhibit increased activities in novel and exploratory environments, in rearing behavior, and deficits in spatial working memory compared to wild-type mice (reviewed in Ref. 50). These mice also exhibited a decrease in methamphetamine-mediated locomotion56 and behavioral sensitization to ethanol but not to amphetamines.56,57 Moreover, the D3R knockout mice exhibit attenuation in the development of tolerance and precipitated withdrawal to chronic morphine administration.58 Studies with D4R knockout mice show that this receptor subtype may be involved in the modulation of novelty-related exploration and avoidance behavior to unconditioned stimuli (reviewed in Ref. 50). With regard to drugs of abuse, these mice are more responsive to the locomotor effects of ethanol, cocaine, or amphetamines than wild-type mice (reviewed in Ref. 50). In addition, D4R alleles associated with novelty seeking behavior have been correlated with drug abuse, attention deficit hyperactivity disorder, and gambling (reviewed in Ref. 50). Studies show that D5R knockout animals exhibit hypertension suggesting that this dopamine receptor subtype may be involved in the regulation of blood pressure (reviewed in Ref. 50). Taken together, these studies reveal a role for dopamine receptors in normal physiology and in drug abuse.
2. RECEPTOR HETEROMERIZATION
In the past decade, the idea that GPCRs associate to form heteromeric complexes with novel pharmacology has emerged (for reviews, see Refs. 59–65). Initial evidence for GPCR heteromerization came from studies showing that activation of a GPCR modulated the function of another GPCR.66–70 The possibility that GPCRs could form heteromers was supported by elegant complementation studies. These included studies using chimeras of the α2c-adrenergic and m3 muscarinic receptors (α2c/m3 and m3/α2c) that, when expressed individually, could not bind to either radiolabeled adrenergic or muscarinic ligands but when coexpressed could.71 Similarly, coexpression of two different mutants of the angiotensin AT1 receptor that individually could not bind to angiotensin led to the restoration of angiotensin binding upon coexpression of these mutant receptors.72
The first direct evidence for GPCR heteromerization came from studies with the metabotropic GABAB receptors. The expression of functional GABAB receptors at the cell surface requires the coexpression of both the GABAB1 and GABAB2 protein subunits.73–76 Another example for the requirement for heteromerization for receptor function is provided by taste receptors where associations between T1R1 and T1R2 form a receptor that recognizes sweet tasting molecules, while those between T1R1 and T1R3 recognize “umami” tasting molecules.77–79
The availability of epitope-tagged GPCRs for immunoisolation of interacting complexes to examine whether two GPCRs are in close proximity for direct interaction has shown that a large number of GPCRs can form heteromers (for reviews, see Refs. 59–61). Scientists in the field have applied the following criteria in order to ascertain that two GPCRs form heteromers: (i) colocalization of the two interacting receptors in the same cellular and subcellular compartment; (ii) immunoisolation of interacting receptors, using receptor-selective antibodies, from wild-type animal tissue that expresses both receptors (but not from animals lacking one of the receptors); (iii) proximity-based assays showing that the two receptors are in close proximity to interact only in cells/tissues expressing both receptors and not in cells/tissues lacking individual receptors; and (iv) unique pharmacological properties as determined using highly selective reagents _targeting the heteromer.
2.1. Methods to study GPCR heteromerization
2.1.1 Techniques to study GPCR heteromerization in vitro
Coimmunoprecipitation assays: In this technique, interacting complexes in tissues are detected using antibodies selective for individual protomers. Alternatively, cells are cotransfected with the GPCRs of interest, each of which is differentially epitope tagged (e.g., with a Flag or myc epitope) and antibodies to the epitope tags used for the assay. Tissues or cells are then solubilized with a buffer-containing detergent, and antibodies to an individual receptor protomer or to the epitope tag on one of the receptors (e.g., the Flag epitope) are used to immunoprecipitate that receptor.80,81 The immunoprecipitates are subjected to Western blot analysis and probed for the presence of the second receptor using antibodies selective for that receptor or to the epitope tag (e.g., the myc epitope) present on the receptor.80,81 The detection of a signal on Western blots indicates that the two receptors form interacting complexes. Given the hydrophobic nature of GPCRs, appropriate controls are used in order to rule out the artifactual detection of interacting complexes. These include carrying out immunoprecipitation studies with tissue from animals lacking one receptor or with cells that express only one receptor, mixing cells that individually express each receptor prior to solubilization and immunoprecipitation, using different detergents for solubilization and/or using cross-linking reagents to cross-link cell surface proteins prior to solubilization and immunoprecipitation.80,81 This strategy was first used to demonstrate opioid receptor heteromers in tissues7 or in cells that coexpress both receptors.82
Bioluminescence resonance energy transfer assay: In order for two receptors to form heteromers, they have to be in close proximity to interact in live cells. One of the earliest proximity-based assays used to examine GPCR heteromerization was bioluminescence resonance energy transfer (BRET) (reviewed in Ref. 83). In this assay, the C-terminus of one of the receptors is fused to Renilla reniformis luciferase (Rluc), while the C-terminus of the other receptor is fused to a genetic variant of green fluorescent protein (GFP) such as yellow fluorescent protein (YFP) (reviewed in Ref. 83). Cells are transfected with both receptors and then treated with the luciferase substrate, coelenterazine h. The action of luciferase on its substrate leads to the emission of light at 480 nm which can excite YFP which then emits light at 530 nm only if the two receptors are less than 100 Å apart (reviewed in Ref. 83). An advantage of this technique is that it does not require conventional light illumination, and this eliminates artifactual results due to autofluorescence (reviewed in Ref. 83). BRET assay was used to demonstrate proximity between opioid receptor types as well as between opioid and other receptors (Table 9.1). Several generations of BRET have been developed. BRET1 uses YFP or enhanced GFP in combination with Rluc and coelenterazine h and has the advantage that the BRET signal can be detected for up to 1 h (reviewed in Ref. 83). BRET2 uses a GFP variant that is excited at ~400 nm and DeepBlueC as the Rluc substrate. This allows significant separation between the excitation and emission spectra of GFP. The BRET2 signal decays rapidly; however, the use of Rluc variants, Rluc2 or Rluc8, ensures a high BRET signal that can last for hours depending on the donor and acceptor combinations chosen (reviewed in Ref. 83). BRET3 makes use of RLuc8 and a variant of red fluorescent protein, mOrange, as donor and acceptor pairs. BRET3 exhibits the most red-shifted emission wavelength (564 nm), and consequently yields a several-fold improvement in light output compared with other BRET assays (reviewed in Ref. 83).
-
Fluorescence resonance energy transfer assay: Another technique commonly used to determine close proximity between two GPCRs is fluorescence resonance energy transfer (FRET). Here, cells are cotransfected with pairs of receptors of interest; one of the receptors is fused to a fluorescent donor and the other to a fluorescent acceptor (reviewed in Ref. 120). The most commonly used fluorescent donor is cyan fluorescent protein (CFP), while the fluorescent acceptor is YFP (reviewed in Ref. 120). Light excitation at 400 nm causes CFP to emit light at 480 nm which can then excite YFP, if it is in sufficiently close proximity, causing it to emit light at 580 nm (reviewed in Ref. 120). Given that the FRET efficiency is inversely proportional to the sixth power of the distance between the two fluorophores, a FRET signal is observed only if the donor–acceptor pair is less than 100 Å apart (reviewed in Ref. 120). An advantage of FRET is that it is amenable to confocal microscopy. Problems with FRET leading to poor signal-to-noise ratio include (i) cross talk, wherein light used to excite the donor also excites the acceptor fluorophore; (ii) bleed through, due to partial overlap between the spectra of the donor and acceptor fluorescent proteins; and (iii) photobleaching (reviewed in Ref. 120). Several modifications of the FRET technique have been developed to improve its signal-to-noise ratio including photobleaching FRET (pbFRET), photoquenching FRET, fluorescence lifetime imaging microscopy (FLIM), and time-resolved FRET. Several excellent reviews describe these techniques in detail.120–124
Several techniques to label GPCRs for FRET studies have been developed including (i) labeling one receptor with a FlAsH tag (a 15-amino acid polypeptide tag with a tetracysteine core (CCXXCC) that covalently binds to a fluorogenic bisarsenical fluorescein ligand whose fluorescence increases upon binding to the polypeptide tag) and fusing the other receptor with CFP, (ii) using mTurquoise instead of CFP for FLIM experiments, (iii) using Eu3+- and Tb3+ cryptate in combination with a fluorescent protein, and (iv) using SNAP- and CLIP tags (reviewed in Ref. 124). Moreover, FRET sensors have been developed to examine ligand binding to a GPCR, receptor activation, conformational changes in receptor dimers, receptor trafficking, and receptor interaction with signaling molecules (reviewed in Ref. 124).
-
Protein complementation: In recent years, protein-fragment complementation assays have been used to examine GPCR heteromerization in living cells. In this approach, cells are transfected with one GPCR fused to a fragment of luciferase and another GPCR fused to a complementary fragment (reviewed in Ref. 121). Reconstitution of active luciferase enzyme would occur only if the two receptors were in close proximity. This approach is known as bimolecular luminescence complementation (BiLC) (reviewed in Ref. 121). A combination of protein complementation with the BRET assay125 has proven to be amenable to the development of sensors to study not only receptor heteromerization but also conformational changes following receptor activation, receptor interactions with signaling molecules, and how the latter are modulated by ligand treatment (reviewed in Ref. 83). Moreover, recent advances in BRET technology have enabled these assays to be carried out in live animals.126
Another protein complementation approach uses a receptor fused with a fragment of YFP (nonfluorescent) and another receptor fused with a complementary YFP fragment (nonfluorescent). YFP fluorescence is reconstituted only when the two receptors are in close proximity. This approach is known as bimolecular fluorescence complementation (BiFC) (reviewed in Ref. 121). The advantage of BiFC over BiLC is that it can provide information about the subcellular localization of receptor interactions (reviewed in Ref. 121). Moreover, a combination of BiFC or BiLC with either BRET or FRET has been used to demonstrate the presence of GPCR trimers or tetramers.127–130
Atomic force microscopy and crystallography: Biophysical approaches such as atomic force microscopy (AFM) and X-ray crystallography have been useful in demonstrating the existence of GPCR dimers. AFM uses a scanning tunneling microscope to measure the movement of a cantilever beam over the sample surface.131 Using this approach, studies showed that rhodopsin, the prototypical family A GPCR, was arranged in dimeric arrays.132 X-Ray crystallography, a method used for determining the arrangement of atoms within a molecule, can provide information about the structural organization of a GPCR. Until very recently, the high-resolution crystal structures for only a few GPCRs were available. These included the structures for rhodopsin and extracellular domains of metabotropic glutamate receptors, which indicated that these receptors could exist as dimers or higher-order structures.133,134 The obstacles in obtaining high-resolution structures for most GPCRs included low expression levels in endogenous tissues, difficulties in expressing the receptor at high enough levels in heterologous cells, structural instability following detergent solubilization, as well as multiple receptor conformational states. Recent approaches aimed at stabilizing a GPCR into a single conformational state, through the use of either monoclonal antibodies that stabilize the conformation of intracellular loop 3 or by replacing a portion of this loop with T4-lysozyme, together with crystallization in the presence of a receptor antagonist, have permitted the determination of high-resolution structures for a number of GPCRs including those for opioid receptors.135–142 In this context, it is to be noted that the recent MOR and chemokine receptor, CXCR4, crystal structures reveal a dimeric receptor arrangement consistent with the idea that these receptors could exist as higher-order structures in vivo.137,139
Table 9.1.
Heteromer pair | Detection techniques | Changes in heteromer properties | References | ||
---|---|---|---|---|---|
Binding | Signaling | Trafficking | |||
DOR–α2AAR | Co-IP, BRET | Not reported | Synergy in inhibition of neuropeptide release; ↑ DOR-mediated neurite outgrowth | Not reported | 84,85 |
DOR–β2AR | Co-IP, BRET | Not changed | Not changed | β2AR internalized by DOR agonists | 86,87 |
DOR–CB1R | Co-IP, BRET | Not reported | ↓ CB1R-mediated signaling | Change in CB1R localization | 88,89 |
DOR–CXCR4 | Co-IP, FRET | Not changed | Nonfunctional | Not changed | 90 |
DOR–D1R | Immuno-EM | Not reported | ↓ DOR signaling by D1R agonists | DOR redistribution by cocaine withdrawal | 91,92 |
DOR–KOR | Co-IP, BRET | ↓ Affinity for selective DOR or KOR ligands; binding cooperativity with DOR+KOR ligands | ↑ With a combination of DOR +KOR agonists | Altered DOR trafficking | 82,86 |
DOR–MOR | Co-IP, BRET | ↓ Affinity for receptor-selective agonists; allosteric modulation of MOR binding by DOR ligands and vice versa | Switch from Gαi to Gαz; heteromer coupling to β-arrestin; potentiation of MOR binding by DOR ligands and vice versa | MOR and DOR endocytosed independently of each other; some DOR agonists induce heteromer internalization | 7,93–103 |
DOR–SNSR | BRET | Not reported | Preferential Gαq signaling | Not reported | 104 |
KOR–β2AR | Co-IP | Not changed | ↓ β2AR signaling | Altered | 105 |
KOR–APJ | Co-IP, BRET | Not reported | ↑ By agonists to either receptor | Not reported | 106 |
MOR–α2AAR | Co-IP, BRET, FRET | Not reported | α2AR ↑ MOR signaling; α2AR ligands ↓ MOR signaling | Agonist to one receptor does not cause heteromer internalization | 107–109 |
MOR–CB1R | Co-IP, BRET, FRET | Not reported | CB1R ↓ MOR signaling; MOR ↓ CB1R signaling | Not reported | 89,110 |
MOR–CCR5 | Co-IP | Not reported | MOR agonist ↓ CCR5 signaling; CCR5 agonist ↓ MOR signaling | Not reported | 111,112 |
MOR–GRPR | Co-IP | Not reported | MOR-mediated Ca2+ signaling only in cells expressing the heteromer | MOR-mediated GRPR internalization | 113 |
MOR–KOR | Co-IP, BRET | ↓ Affinity for MOR-selective ligands | Not reported | Not reported | 82,114,115 |
MOR–NK-1 | Co-IP, BRET | Not changed | Not changed | MOR internalization by NK-1 agonist and vice versa | 116 |
MOR–OFQ/N | Co-IP | ↑ Affinity for MOR-selective ligands | ↓ Potency of MOR ligands | Not reported | 117,118 |
MOR–SSTR2 | Co-IP | Not changed | Not changed | ↑ Heteromer internalization by sSSTR2 agonist | 119 |
2.1.2 Techniques to study GPCR heteromerization in vivo
Ligand-aided FRET: The presence of GPCRs in endogenous tissue can be detected by FRET assays using fluorescently labeled ligands to each receptor protomer. Since both agonists and antagonists can be used in such studies, they provide additional information on the conformational state of the receptor.143 This approach makes use of fluorophores compatible with TR-FRET to label the ligands in order to obtain a high signal-to-noise ratio.143 Thus, ligands are labeled with donor–acceptor pairs (typical examples of donor–acceptor pairs optimal for TR-FRET include Eu-PBBP, Lumi4-Tb, or Alex-64-d1), and the assay is then carried out in tissue patches.143 Such an approach was used to detect the presence of oxytocin receptor dimers/oligomers in mammalian gland.143
Antibody-aided FRET: The presence of GPCR heteromers in live cells can be detected by a combination of selective antibodies and FRET. For example, in live cells expressing epitope-tagged receptors, secondary antibodies labeled with either fluorescein or rhodamine were used in pbFRET experiments to monitor the formation of SSTR1–SSTR5 and D2R–SSTR5 heteromers.144,145 In another study, antibodies to epitope tags on the receptor were labeled with either Eu3+ or allophycocyanin protein and used in TR-FRET experiments to demonstrate the presence of DOR oligomers at the cell surface.146 An extension of the latter approach could be used to examine GPCR heteromerization in endogenous tissue by labeling monoclonal antibodies that are highly selective to individual receptors in the heteromer with complementary TR-FRET donor–acceptor pairs. Such an approach was used to detect the presence of GABAB receptor tetramers on brain membranes.147
Proximity-based ligation assays (PLAs): Recently, PLAs have been used to detect the presence of GPCR heteromers in endogenous tissue.148,149 This is an antibody-based approach where tissue sections of interest are first incubated with anti-GPCR selective antibodies.148,149 The antibody to each protomer in the heteromeric complex under investigation is raised in a different species (rabbit or mouse). Sections are then incubated with species-specific secondary antibodies coupled to complementary oligonucleotide sequences (PLA probes).148,149 When the antibodies to the two receptors are in close proximity (<17 nm), the PLA probes can be ligated using ligase and a ligation solution comprising of two oligonucleotides that will hybridize with the two PLA probes to form a closed circle.148,149 The ligated closed circle is then amplified by polymerase in a rolling-circle amplification reaction in the presence of fluorescently labeled oligonucleotides.148,149 Hybridization of the fluorescently labeled nucleotides appears as afluorescent spot that can then be visualized by fluorescence microscopy.148,149 This assay has been used to detect the presence of CB1R–CB2R receptor heteromers in the pineal body and nucleus accumbens and of dopamine D2–adenosine A2A receptor heteromers in the striatum.148,149
Heteromer-selective antibodies: Monoclonal antibodies that selectively recognize a GPCR heteromer are useful tools that can be used to not only determine the tissue distribution of the heteromer but also its possible physiological role. A subtractive immunization strategy has been described for the development of such antibodies.7,9,10,88 In this approach, mice are first made tolerant to nonspecific cell membrane epitopes by treatment with cyclophosphamide.7,9,10,88 Mice are then immunized with membranes from cells expressing the heteromer of interest.7,9,10,88 Antibodies selective for MOR–DOR, DOR–CB1R, or DOR–AT1R heteromers have been generated using this approach.7,9,10,88 Studies using these antibodies show that the antibodies are able to selectively block signaling by specific heteromers and that MOR–DOR heteromers are increased in brain regions involved in pain processing under conditions leading to the development of tolerance to morphine, while DOR–CB1R heteromer levels are increased in specific brain regions during neuropathic pain, and DOR–AT1R heteromer levels are increased during ethanol-induced liver fibrosis.7,9,10,88
2.2. Opioid receptor heteromerization
Early radiolabeled binding studies, as well as studies measuring the antinociceptive effects of selective opioid receptor ligands, suggested the presence of multiple opioid receptor subtypes.150,151 However, to date, the cDNAs for only MOR, DOR, and KOR have been identified.2,20 This suggested the possibility that the multiple opioid receptor subtypes detected by binding and antinociceptive studies could arise from the formation of heteromeric complexes between opioid receptor subtypes (MOR, DOR, KOR) or between the latter and other GPCRs. The availability of cDNAs for epitope-tagged receptors together with coimmunoprecipitation and/or proximity-based assays has enabled studies showing that opioid receptors can form heteromers with a number of GPCRs (see Table 9.1). These are described below.
DOR–KOR heteromers
The first opioid receptor heteromer to be detected and characterized involved DOR and KOR.82 Coimmunoprecipitation studies, using differentially epitope-tagged receptors, showed that DOR and KOR could form interacting complexes.82 BRET studies showed that both receptors were in close enough proximity (<100 Å) in live cells to directly interact.86 Analysis of the binding, signaling, and trafficking properties of DOR–KOR heteromers revealed that the latter exhibited properties distinct from that of the individual receptors. For example, the binding affinities of selective DOR and KOR ligands were reduced in the DOR–KOR heteromer; however, a combination of DOR and KOR ligands exhibited cooperativity in binding that in the case of agonists was translated into a potentiation in signaling responses.82 This binding cooperativity was also observed with a combination of DOR and KOR antagonists.82 Bivalent ligands consisting of DOR and KOR antagonists (naltrindole and 5′-guanidinonaltrindole (5′-GNTI), respectively) linked by spacers of variable length were used to _target the DOR–KOR heteromer.152 One of these bivalent ligands, KDN-21, exhibited a higher affinity and selectivity for DOR–KOR heteromers compared to DOR and KOR homomers.153 Pharmacological and binding data indicate that KDN-21 binds to DOR–KOR heteromers of the DOR1–KOR2 phenotype.152,153 Interestingly, a study identified 6′-guanidinonaltrindole (6′-GNTI) as a DOR–KOR heteromer-selective agonist that induced antinociception when administered to the spinal cord but not to the brain.154 However, a recent study has suggested that 6′-GNTI is a KOR-biased ligand155; thus, it remains to be seen if 6′-GNTI represents a DOR–KOR heteromer-biased ligand. Studies that suggest a role for DOR–KOR heteromers in pain pathways are described in Section 3.
DOR–MOR heteromers
This is the most extensively studied opioid receptor heteromer due to its role in morphine-mediated antinociception and in the development of tolerance and dependence to opiates. A comprehensive body of evidence suggested the possibility of DOR–MOR heteromerization (reviewed in Ref. 156). Noteworthy are radiolabeled studies indicating that (i) there is an allosteric modulation of radiolabeled binding to DOR by MOR ligands157,158; (ii) intracerebroventricular potentiation of morphine-mediated antinociception by nonantinociceptive doses of DOR agonists which can be blocked by the DOR antagonists ICI174,864 and naltrindole-5′-isothiocyanate159; (iii) potentiation of acute morphine-mediated antinociception by DOR antagonists and inhibition of development of tolerance or reduction of already developed tolerance to morphine160; (iv) prevention of the development of tolerance and dependence in mice by pretreatment with DOR antagonists161; and (v) lack of development of tolerance to morphine in DOR knockout mice.162
Evidence that MOR and DOR form heteromeric complexes in vitro came from coimmunoprecipitation and BRET studies using differentially epitope-tagged receptors.93–95 In addition, coimmunoprecipitation studies using receptor-selective antibodies revealed the presence of DOR–MOR heteromers in spinal cord membranes from wild-type but not DOR knockout mice.94 More recently, antibodies that selectively recognize DOR–MOR heteromers were used to detect the presence of these heteromers in brain regions of wild-type but not MOR or DOR knockout mice.7
DOR–MOR heteromers exhibit distinct pharmacological properties from that of individual receptors.93–95 Low nonsignaling doses of DOR or MOR ligands potentiate the binding and signaling of MOR or DOR receptors, respectively.93,94,96 Moreover, the potency of highly selective synthetic MOR or DOR ligands is decreased, while the affinity for endomorphin-1 and DOR-selective agonists is increased in DOR–MOR heteromers as compared to MOR or DOR.95,97
Studies show that DOR–MOR heteromerization leads to changes in the signaling pathways activated by the heteromers compared to individual receptor homomers. In this context, a study showed that the DOR–MOR heteromer associates with pertussis-insensitive Gαz instead of the pertussis-sensitive Gαi and this interaction occurs in the endoplasmic reticulum.98,99 Another study reported that activation of a protomer in the DOR–MOR heteromer leads to a switch in signaling from Gαi- (as it occurs in receptor homomers) to β-arrestin-mediated signal transduction leading to changes in the spatiotemporal dynamics of ERK1/2 phosphorylation and ultimately differential activation of transcription factors.100 It has also been reported that while the MOR-selective agonist, DAMGO, stimulates Ca2+ signaling in cells expressing DOR–MOR heteromers by activating phospholipase C and releasing Ca2+ from intracellular stores, it inhibits Ca2+ signaling in GH3 cells expressing only MOR by inhibiting adenylyl cyclase activity and voltage-gated Ca2+ channels and by activating inwardly rectifying K137+. Taken together, these studies indicate that heteromerization increases the repertoire of signaling for opioid receptors.
A few studies have examined how DOR–MOR heteromerization modulates the trafficking of individual receptors. One study reported that treatment with receptor-selective agonists (DAMGO or deltorphin II but not DPDPE or DSLET) led to internalization of the DOR–MOR heteromer.99 However, another study examining receptor turnover in cells that expressed increasing levels of DOR with constant levels of MOR reported that the two receptors endocytosed independently of each other in the context of the heteromer.102 Finally, a recent study reported that methadone could promote the internalization of the DOR–MOR heteromer _targeting it to the degradation machinery.103 Together, these data suggest that the internalization of the DOR–MOR heteromer could be a ligand type-dependent event. Recent studies suggest a role for these heteromers in pathways leading to development of tolerance to morphine; this is discussed in Section 3.
DOR–CB1R heteromers
Early studies examining interactions between DOR and CB1R showed that (i) long-term exposure to a DOR agonist attenuated CB1R-mediated stimulation of G-protein activity in N18TG2 neuroblastoma cells that coexpress DOR and CB1R163; (ii) the DOR antagonist, naltrindole, blocked Δ9-THC-mediated anxiolytic responses105; (iii) CB1R levels as well as agonist-mediated G-protein activity are increased in the substantia nigra of mice lacking DOR as compared to wild-type animals164; and (iv) DOR activity is increased in the caudate putamen of CB1R knockout mice compared to wild-type mice.165
DOR–CB1R heteromers are detected in HEK-293 and in Neuro 2A cells coexpressing both receptors by coimmunoprecipitation and BRET assays.88,89 DOR and CB1R are colocalized in cortical neurons as examined by immunofluorescence studies with receptor-selective antibodies.88 Interestingly, heteromerization leads to a differential recruitment of trafficking (adaptor) proteins; CB1R associates with the adaptor protein AP-3 in the absence of DOR and with AP-2 in the presence of DOR.88 This, in turn, leads to differential localization of CB1R from intracellular vesicles, in the absence of DOR, to the cell surface in the presence of DOR.88 In addition, lentiviral-mediated knockdown of DOR in F11 cells that endogenously coexpress CB1R and DOR leads to a decrease in cell surface localization of CB1R.88 Heteromerization also alters CB1R signaling; the potency of the CB1R agonist, Hu-210, is decreased in cells expressing DOR–CB1R in assays that measure G-protein activity or ERK1/2 phosphorylation, and in these cells, knockdown of DOR leads to an increase in the potency of Hu-210.88 Moreover, DOR–CB1R heteromerization leads to a PLC-mediated recruitment of β-arrestin 3 to the heteromeric complex, and to the activation of novel signaling pathways that promote cell survival.88 Recent studies suggest a role for these heteromers in pain pathways; this is discussed in Section 3.
DOR–CXCR4 heteromers
Given that both DOR and chemokine CXCR4 receptors are expressed in immune cells, coimmunoprecipitation and FRET assays were used to determine whether both receptors could form heteromers.90 These studies showed that DOR and CXCR4 form interacting complexes.90 Moreover, the studies showed that simultaneous activation of DOR and CXCR4 in cells coexpressing both receptors did not affect cell viability, chemokine binding, or receptor internalization but led to the formation of nonfunctional complexes.90 This suggests that DOR–CXCR4 heteromerization provides a means to silence CXCR4 function and could be important in the regulation of immune function.
DOR–sensory neuron-specific receptor heteromers
Sensory neuron-specific receptors (SNSRs) are mainly expressed in dorsal root ganglia in the spinal cord, where they mediate pronociceptive responses.166 Given that DOR and SNSR-4 share localization in dorsal root ganglia, can be activated by the bovine adrenal medulla peptide 22 (BAM22), and their functions are antagonistic to each other, led to studies examining whether both receptors could form heteromeric complexes. BRET assays in HEK-293 cells coexpressing differential epitope-tagged receptors indicated that both receptors were in sufficiently close proximity to directly interact in live cells.104 In cells expressing DOR–SNSR-4 heteromers, activation of SNSR-4 alone led to Gαq-mediated signaling, while activation of DOR alone resulted in Gαi-mediated signaling. However, coactivation of both receptors by receptor-specific agonists led to a preferential signaling via Gαq.104 Interestingly, the endogenous ligands for SNSR are derived from the proteolytic processing of the proenkephalin precursor that also generates the antinociceptive endogenous DOR ligands, Leu- and Met-enkephalin.167 Moreover, one of these peptides, BAM22, has been shown to bind to both DOR and SNSR-4.167 Activation of DOR–SNSR-4 heteromers with BAM22 leads to signaling via Gαq. This is the first demonstration of an endogenous peptide as a bivalent agonist of a GPCR heteromer.
DOR–β2 adrenergic receptor heteromers
Interactions between DOR, a Gαi-coupled receptor, and the β2 adrenergic receptor (β2AR), a Gαs-coupled receptor, were suggested by studies showing that in the isolated isovolumic rat heart the DOR agonist, Leu-enkephalin, significantly inhibited β2AR-mediated positive ionotropic effects and stimulation of cAMP levels, effects that were blocked by the opioid antagonist, naloxone, and by pertussis toxin treatment.168 Moreover, in isolated rat ventricular myocytes, nanomolar concentrations of Leu-enkephalin could block β2AR-mediated increases in sarcolemmal L-type Ca2+ currents, cytosolic Ca2+ transients, and contraction.169 Coimmunoprecipitation and BRET assays using differentially epitope-tagged receptors demonstrated the formation of DOR–β2AR heteromers in HEK-293 cells.86,87 While the binding and signaling properties of individual protomers in the DOR–β2AR heteromer were not changed by heteromerization, individual receptor trafficking was, in that DOR agonists could internalize β2AR only in cells coexpressing both receptors.87
DOR–α2A adrenergic receptor heteromers
A number of studies have suggested interactions between DOR and α2A adrenergic receptors (α2AAR). These include studies showing that (i) a selective imidazole/α2AR agonist synergizes with the DOR agonist, deltorphin II, to block substance P elicited nociception170; (ii) the synergy between agonists for the two receptors was observed in α2CAR and in MOR knockout mice but not in α2AAR knockout mice suggesting an involvement of α2AAR and of DOR in these interactions171–173; and (iii) cotreatment of synaptosomes isolated from the spinal cord or of spinal slices with α2AAR and DOR agonists led to potentiation of inhibition of K+-stimulated release of neuropeptides.84,174
Studies using differentially epitope-tagged receptors expressed in heterologous cells together with coimmunoprecipitation and BRET assays show that α2AAR and DOR are in close proximity (<100 Å apart) to form interacting complexes.85 Not much is known about how DOR–α2AAR heteromerization modulates the pharmacological, signaling, and trafficking properties of this heteromer pair. One study suggested the involvement of protein kinase C in heteromer-mediated synergy in inhibition of calcitonin gene-related peptide release from spinal cord slices,84 while another indicated that the presence of α2AAR is sufficient to potentiate DOR-mediated neurite outgrowth in Neuro 2A cells.85 The latter study indicated that just the presence of α2AAR is sufficient to modulate DOR activity; whether the converse also occurs and how ligand occupancy of both protomers in the DOR–α2AAR heteromer modulates individual receptor function need to be further evaluated.
DOR–D1R heteromers
Interactions between DOR and D1R were suggested by studies showing (i) that alterations in dopaminergic neurotransmission modulate the endogenous levels of opioid peptides175–177; (ii) attenuation of DOR-mediated signaling by the D1R agonist, SKF 82958178; and (iii) enhancement of climbing behavior in mice by a combination of DOR and D1R agonists.179
Immunoelectron microscopy detected the presence of DOR and D1R immunoreactivity in a subset of dendrites in the dorsolateral striatum and nucleus accumbens of rats, indicating that both receptors were present in the same neuron.91,92 Moreover, withdrawal from cocaine, a drug that blocks dopamine transporter activity and thereby alters dopamine neurotransmission by increasing and prolonging signaling by postsynaptic dopamine receptors caused a redistribution of DOR in the nucleus accumbens.91 Although these colocalization studies indicate that DOR–D1R could form heteromers, this needs to be further evaluated by studies using epitope-tagged receptors, coimmunoprecipitation, and proximity-based assays.
MOR–KOR heteromers
Very few studies have examined heteromerization between MOR and KOR. An early study using coimmunoprecipitation of differentially epitope-tagged receptors did not detect the presence of interacting MOR–KOR complexes.82 This does not rule out the formation of MOR–KOR heteromers if the latter are not stable under the detergent conditions used in the coimmunoprecipitation studies. Another study used BRET to show that MOR and KOR are in close proximity to interact in live cells.114 A recent study reported the formation of MOR–KOR heteromers in spinal cord membranes of female, but not male, rats and showed that it is dependent on the stage of the estrous cycle.115 Interestingly, this study identified dynorphin 1–17 as an MOR–KOR heteromer-selective ligand.115
MOR–α2A adrenergic heteromers
Studies describing synergistic interactions between the antinociceptive effects of MOR and α2AAR agonists180,181 and with α2AAR knockout mice that exhibit a reduction in the analgesic potency of morphine173 suggested the possibility of MOR–α2AAR heteromerization.
Differential epitope-tagged receptors together with coimmunoprecipitation, BRET, or FRET assays demonstrated the formation of MOR–α2AAR heteromers.107,108 Studies examining heteromer-mediated signaling showed that while the presence of α2AAR is sufficient to enhance MOR signaling, coactivation of both receptors in the heteromeric complex leads to a significant attenuation of signaling responses.107 Proximity-based assays (FRET, BRET) showed that, in cells coexpressing MOR–α2AAR heteromers, treatment with morphine induces conformational changes in norepinephrine occupied α2AAR leading to a decrease in Gαi-mediated signaling.108
Finally, a study examining the trafficking of MOR–α2AAR heteromers showed that treatment with the agonist to one receptor protomer did not lead to cointernalization of the heteromeric partner receptor.109 However, given that, in addition to heteromers, the cells express individual receptor homomers, gives rise to the possibility that the agonists are in fact causing internalization of the homomer rather than the _targeted receptor in the heteromer. Thus further studies are required to evaluate the trafficking properties of MOR–α2AAR heteromers.
MOR–CB1R heteromers
Several studies have described functional interactions between MOR and CB1R (reviewed in Ref. 43). These include studies showing that (i) Δ9-THC enhances the potency of morphine in animal models4; (ii) conditioned place preference to Δ9-THC is not observed in MOR knockout mice17; and (iii) CB1R knockout mice exhibit a reduction in the reinforcing properties of morphine as well as in the severity of withdrawal symptoms to this opioid.182
Coimmunoprecipitation, BRET, and FRET assays using differentially epitope-tagged receptors detected the presence of MOR–CB1R heteromers in heterologous expressing systems.89,110 While the presence of unliganded CB1R does not affect MOR-mediated signaling, ligand binding to CB1R decreases MOR-mediated signaling.89 Conversely, ligand binding to MOR decreases CB1R-mediated signaling.89 These interactions had physiological relevance as observed by inhibition of neurite outgrowth in Neuro2A cells that coexpress MOR and CB1R following cotreatment with agonists to both receptors.89 Electrophysiological studies with oocytes coexpressing MOR and CB1R and where one of the receptors is fused to a chimeric Gqi5 showed that agonists to either receptor elicited Ca2+-activated Cl− currents thus demonstrating the formation of a functional receptor heteromer.110 However, this study did not examine the effect of coadministration of agonists to both receptors on Ca2+-activated Cl− currents.
MOR–chemokine receptor 5 heteromers
Studies have described the presence of MOR in immune cells and that receptor activation by morphine increases the expression of chemokine receptor 5 (CCR5) suggesting interactions between these two receptors.183,184 Coimmunoprecipitation studies using differentially epitope-tagged receptors showed that MOR–CCR5 interacting complexes could be detected only in cells coexpressing both receptors.111,112 Examination of the properties of the MOR–CCR5 heteromer showed that treatment with either the MOR agonist, DAMGO, or the CCR5 agonist, RANTES, induced chemotaxis in cells coexpressing both receptors.111 However, pretreatment with DAMGO inhibited RANTES-induced chemotaxis, increased CCR5 phosphorylation, and decreased G-protein coupling to CCR5.111 The converse was observed when cells were pretreated with RANTES111 suggesting cross-desensitization of MOR and CCR5 within the heteromeric complex.
MOR–gastrin-releasing peptide receptor heteromers
Studies showing that (i) MOR agonist administration induces itching (also known as pruritus)185–187; (ii) scratching responses to itch-inducing stimuli including morphine administration are abrogated in mice lacking gastrin-releasing peptide receptor (GRPR)113,188,189; and (iii) colocalization of MOR and GRPR in the dorsal horn of the spinal cord113 led to the examination of interactions between the MOR isoform, MOR1D (comprising exons 1–3 and 8–9 of the Oprm gene) and GRPR.113
Direct interactions between MOR1D and GRPR were suggested by coimmunoprecipitation studies carried out in heterologous cells expressing epitope-tagged receptors or with spinal cord membranes that endogenously express both receptors.113 Examination of the signaling properties of the MOR1D–GRPR heteromer showed that morphine-induced intracellular Ca2+ release only in cells coexpressing both receptors.113 Interestingly, treatment with morphine enhanced GRPR internalization, while treatment with the GRPR agonist had no effect on MOR internalization in cells coexpressing both receptors.113
MOR–orphanin FQ/nociceptin receptor heteromers
Interactions between the orphanin FQ/nociceptin receptor (OFQ/N) and MOR have also been described. These include studies showing that an OFQ/N receptor antagonist potentiated the analgesic effects of morphine both in naïve mice and in mice that were tolerant to this opioid.190,191 Coimmunoprecipitation studies using differentially epitope-tagged receptors showed that MOR and OFQ/N receptors could form interacting complexes in cells coexpressing both receptors.117,118 MOR–OFQ/N heteromers exhibited an enhanced affinity for MOR-selective ligands.117 Interestingly, MOR–OFQ/N receptor heteromerization attenuated the potency of MOR-mediated inhibition of adenylyl cyclase activity and ERK1/2 phosphorylation.118 Moreover, treatment with the OFQ/N receptor agonist, nociceptin, led to desensitization of both MOR- and OFQ/N receptor-mediated signaling, while treatment with the MOR agonist, DAMGO, desensitized only MOR-mediated signaling.118 These studies suggest that MOR–OFQ/N receptor heteromerization may play a role in modulating MOR receptor function.
MOR–NK-1 receptor heteromers
Substance P exerts its effects through the activation of neurokinin-1 (NK-1) receptors. Both MOR and NK-1 receptors have been detected in dorsal horn neurons of the spinal cord as well as in brain regions implicated in mood disorders, stress responses as well as those that mediate the rewarding effects of drugs of abuse.192,193 In addition, treatment with morphine leads to an increase in the expression of NK-1 receptors in cortical neurons leading to a greater substance P-mediated Ca2+ mobilization than in untreated neurons.194 Moreover, activation of NK-1 receptors modulates neuronal morphine-mediated endocytosis and MOR desensitization.195 These observations together with those showing that the rewarding effects of morphine are not present in NK-1 receptor knockout mice led to studies examining whether these two receptors formed heteromeric complexes.196 Coimmunoprecipitation and BRET assays using differentially epitope-tagged receptors demonstrate the presence of MOR–NK-1 receptor heteromers in HEK-293 cells coexpressing both receptors.116 MOR–NK-1 receptor heteromerization did not lead to changes in the binding and signaling properties of individual protomers.116 However, treatment with the MOR-selective agonist, DAMGO, led to phosphorylation and cointernalization of NK-1 receptors, while treatment with substance P induced the phosphorylation and cointernalization of MOR.116 Moreover, in cells coexpressing MOR and NK-1 receptors but not in cells expressing only MOR, cotreatment with substance P and DAMGO-induced cointernalization of both receptors along with β-arrestin into the same endosomal compartment leading to a delay in the recycling and resensitization kinetics of MOR.116
MOR–somatostatin 2A receptor heteromers
Another receptor pair investigated for the possible formation of heteromeric complexes involved MOR and somatostatin 2A receptors (sst2a) based on the fact that the latter exhibited 38% homology with the amino acid sequence of MOR.119 Coimmunoprecipitation studies using differentially epitope-tagged receptors demonstrated the presence of interacting MOR–sst2a receptor complexes in HEK-293 cells coexpressing both receptors.119 MOR–sst2a receptor heteromerization did not affect protomer-mediated ligand binding and signaling; however, it modulated agonist-mediated receptor trafficking.119 In this context, in cells coexpressing MOR and sst2a, the sst2a receptor agonist promoted the internalization of both receptors, while the MOR agonist promoted only the internalization of MOR.119 Although it is possible that MOR–sst2a heteromers do not undergo internalization following treatment with MOR agonists (the observed internalization being that of MOR homomers), it is to be noted that MOR agonists desensitize sst2a receptor signaling and sst2a receptor agonists desensitize MOR-mediated signaling.119
KOR–β2AR heteromers
Both KOR and β2AR are present in the heart and treatment with the β2AR agonist, isoproterenol, has been shown to increase radiolabeled U69,593 binding to KOR present in rat cardiac sarcolemma.197,198 This suggested interactions between KOR and β2AR. Coimmunoprecipitation studies using differentially epitope-tagged receptors expressed in HEK-293 cells indicated that KOR and β2AR formed interacting complexes.87 While KOR–β2AR heteromerization did not affect the pharmacology of each protomer in the complex, it did modulate trafficking and signaling as examined by observing the phosphorylation of ERK1/2 in cells coexpressing both receptors.87 For example, treatment with a β2AR agonist reduced β2AR internalization in cells coexpressing KOR–β2AR, but could induce internalization in cells expressing only β2AR.87 Moreover, β2AR-mediated increases in ERK1/2 phosphorylation are significantly attenuated in cells coexpressing KOR–β2AR heteromers. Together, these results indicate an important physiological role for KOR–β2AR heteromerization in modulation of β2AR function.
KOR–apelin receptor heteromers
The apelin receptor (APJ) binds to the apelin peptide and plays a role in the modulation of several functions including cardiovascular functions, neuroprotection, pain perception, and regulation of food and water intake.199,200 Given that both KOR and APJ exhibit a similar distribution and function in the cardiovascular and nervous system, studies examined the possible heteromerization between these two receptors.106 Coimmunoprecipitation, colocalization, and BRET assays show that KOR and APJ can form interacting complexes in HEK-293 cells.106 KOR–APJ heteromerization leads to potentiation of ERK1/2 phosphorylation following treatment with either the APJ or KOR agonist compared to what is observed in cells expressing only homomers for either receptor.106 Moreover, KOR–APJ heteromerization leads to a decrease in the activity of protein kinase A, an increase in the activity of protein kinase C, and an increase in cell proliferation.106
2.3. Cannabinoid receptor heteromerization
Compared to opioid receptors, fewer studies have examined heteromerization of cannabinoid receptors. While heteromerization between opioid receptor subtypes and CB1R has been described in Section 2.1, in this section we describe cannabinoid receptor heteromers that do not involve opioid receptors (see Table 9.2).
Table 9.2.
Heteromer pair | Detection techniques | Changes in heteromer properties | References | ||
---|---|---|---|---|---|
Binding | Signaling | Trafficking | |||
CB1R–A2AR | Co-IP, BRET | Not reported | CB1R antagonist ↓ A2AR signaling; A2AR antagonist ↓ CB1R signaling | Not reported | 201 |
CB1R–β2AR | BRET | Not reported | Alteration in signaling of individual protomers | CB1R internalization by β2AR agonist | 202 |
CB1R–AT1R | Co-IP, BRET | Not reported | AT1R signals via Gαi instead of Gαq; CB1R agonists ↑ AT1R signaling; CB1R antagonists ↓ AT1R signaling | Not reported | 10 |
CB1R–CB2R | BRET, proximity ligation assay | Not reported | Antagonistic interactions | Not reported | 148 |
CB1R–D2R | Co-IP | Not reported | CB1R signals via Gαs in the presence of D2R agonist | Not reported | 203–206 |
CB1R–OX1R | Co-IP, TR-FRET | Not reported | CB1R ↑ OX1R signaling | OX1R agonist ↑ heteromer internalization | 207,208 |
CB1R–D2R–A2AR | BRET+FRET; bimolecular fluorescence complementation+BRET | Not reported | Not reported | Not reported | 129,209 |
This table does not include opioid–cannabinoid receptor heteromers. These are described in Table 9.1.
CB1R–adenosine 2A receptor heteromers
Several studies suggested interactions between CB1R and adenosine 2A receptors (A2AR). These include studies showing that (i) activation of A2AR blocks, while inhibition of A2AR promotes, CB1R-mediated effects on evoked glutamate release210; (ii) the rewarding and aversive effects of Δ9-THC were significantly reduced in A2AR knockout mice211; (iii) A2AR antagonist treatment blocks the synergy between MOR and CB1R for CRE-mediated gene transcription212; (iv) locomotor behavior induced by administration of A2AR antagonists is attenuated by CB1R antagonists or in CB1R knockout mice213; and (v) low doses of A2AR antagonists attenuated while higher doses potentiated the self-administration of Δ9-THC and anandamide.214
The formation of interacting CB1R–A2AR complexes in rat striatum was detected by coimmunoprecipitation studies using receptor-selective antibodies.201 BRET using differentially epitope receptors demonstrated that CB1R and A2AR are in close proximity to interact in HEK-293 cells.201 Studies carried out in SH-SY5Y neuroblastoma cells, that endogenously express CB1R and A2AR, show that CB1R activation inhibits forskolin-mediated increases in cAMP levels, and that the latter is blocked by an A2AR antagonist.201 Moreover, A2AR-mediated increases in cAMP levels are blocked by a CB1R antagonist.201
CB1R–AT1 angiotensin receptor heteromers
Early studies demonstrating an upregulation of CB1R in liver cells that have resident AT1 angiotensin receptors (AT1R) prompted the examination of interactions between these two receptors.215–218 The heteromerization of CB1R and AT1R was demonstrated by coimmunoprecipitation, colocalization, and BRET assays using differentially epitope-tagged receptors.10 Examination of the signaling properties of the CB1R–AT1R heteromer shows that, in cells coexpressing both receptors, the AT1R agonist induces a rapid, robust dose-dependent increase in ERK1/2 phosphorylation that is drastically reduced either by RNAi-mediated knockdown of CB1R or by inhibiting the activity of diacylglycerol lipase, the enzyme responsible for the synthesis of the endocannabinoid 2-AG.10 In addition, CB1R agonists potentiate while antagonists block AT1R-mediated increases in ERK1/2 phosphorylation.10 Interestingly, an antibody that selectively recognizes the CB1R–AT1R heteromer blocks AT1R-mediated ERK signaling in cells coexpressing both receptors.10 Moreover, in cells expressing the CB1R–AT1R heteromer, CB1R-mediated ERK1/2 phosphorylation is only observed following activation of AT1R by a very low dose of agonist that by itself does not induce ERK1/2 phosphorylation.10
Examination of receptor G-protein coupling shows that, in cells expressing the CB1R–AT1R heteromer, AT1R-mediated ERK1/2 phosphorylation occurs via Gαi instead of Gαq. Moreover, AT1R-mediated intracellular Ca2+ mobilization occurs via Gαq but requires the presence of CB1R since it is significantly attenuated by RNAi-mediated knockdown of CB1R.10 Interestingly, activation of CB1R did not increase intracellular Ca2+ mobilization, although the CB1R agonist could potentiate AT1R-mediated effects.10 Taken together, these studies show that CB1R–AT1R heteromerization provides a mechanism for modulation of AT1R function by CB1R that could be of clinical relevance in pathologies involving these receptors (see Section 3).
CB1R–β2AR heteromers
Based on coexpression of CB1R and β2AR in several tissues and on inhibition of noradrenergic neurotransmission by CB1R a study examined whether both receptors formed interacting complexes.202 BRET assays using differentially epitope-tagged receptors showed that both receptors were in close proximity to form complexes in live cells.202 The BRET signal was increased in the presence of a CB1R antagonist suggesting that the latter either caused a change in receptor conformation (bringing in close proximity the acceptor and donor pairs) or caused an increase in the surface levels of this heteromer.202 Heteromerization between CB1R and β2AR was shown to lead to alterations in the signaling properties of individual receptors in that it led to an increase in CB1R-mediated ERK1/2 phosphorylation and a decrease in CREB phosphorylation compared to cells expressing only CB1R.202 Moreover, β2AR-mediated ERK1/2 phosphorylation was attenuated by an inverse agonist but not by a neutral agonist to CB1R.202 CB1R–β2AR heteromerization modulated the trafficking properties of individual receptors in that treatment with a β2AR receptor agonist induced CB1R internalization in cells expressing both receptors.202
CB1R–CB2R heteromers
A recent report has described heteromerization between CB1R and CB2R in vivo.148 The authors used BRET assays with differentially epitope-tagged receptors in HEK-293 cells to show that CB1R and CB2R are in close proximity in live cells.148 They also carried out a proximity ligation assay in SH-SY5Y neuroblastoma cells (endogenously express CB1R) transfected with epitope-tagged CB2R, in pinealocytes obtained from rat pineal glands, and in rat nucleus accumbens slices to show that both receptors are in close proximity (<17 nm) to form interacting complexes.148 The authors examined the signaling properties of CB1R–CB2R heteromers in SH-SY5Y cells transfected with CB2R and reported that a CB2R agonist induces Akt/PKB phosphorylation only in cells that coexpressed both receptors, and that this is significantly attenuated by cotreatment with the CB1R agonist.148 Interestingly, this translated into a physiological response given that CB2R-mediated neurite outgrowth in cells expressing CB1R–CB2R heteromers was blocked by the CB1R agonist.148 These results suggest that the interactions between CB1R and CB2R are antagonistic in nature.
CB1R–dopamine D2 receptor heteromers
A number of studies have reported interactions between CB1R and D2R. These include studies showing that (i) the D2R agonist, quinpirole, mediated motor behavior is stimulated by the CB1R antagonist, SR141716A203; (ii) G-protein coupling to CB1R is modulated by D2R in cells coexpressing both receptors204,205; and (iii) in primary striatal neuronal cultures treatment either with the CB1R agonist, Hu-210, or with the D2R agonist, quinpirole, leads to inhibition of forskolin-stimulated cAMP levels,204 whereas coadministration of Hu-210 and quinpirole resulted in increases in cAMP levels mediated via CB1R stimulation of Gαs.204 Interestingly, while the presence of D2R is required for CB1R coupling to Gαs, persistent activation of D2R switches the coupling from Gαs to Gαi.205 The formation of CB1R–D2R heteromeric complexes was demonstrated by coimmunoprecipitation studies using differentially epitope-tagged receptors that show that cotreatment with subsaturating concentrations of agonists to both receptors causes an increase in the levels of CB1R–D2R heteromers.206
CB1R–orexin-1 receptor heteromers
Coimmunoprecipitation studies along with homogenous TR-FRET assays using SNAP- and CLIP-tagged substrates were used to show that CB1R and orexin-1 receptors (OX1R) form multimeric complexes.207 Examination of the trafficking properties of this heteromer showed that the OX1R agonist was more efficient at promoting heteromer internalization compared to OX1R internalization.207 In heterologous cells coexpressing CB1R and OX1R, a Gq/11-coupled receptor, treatment with the OX1R agonist led to a potent and long-lasting increase in ERK1/2 phosphorylation compared to cells expressing only OX1R, and that this increase was blocked by a CB1R antagonist or by pertussis toxin pretreatment.208 Moreover, coadministration of CB1R and OX1R agonists did not result in additive effects suggesting that just the presence of CB1R was sufficient to modulate OX1R signaling.208 Given the involvement of CB1R and OX1R in feeding, _targeting this heteromer pair could lead to the identification of novel therapeutics for the treatment of obesity.
CB1R–D2R–A2AR heteromers
Above, we described studies showing heteromerization between CB1R and D2R and between CB1R and A2AR. In addition, D2R–A2AR heteromerization has been reported.219 This led to the hypothesis that the three receptors could associate to form an oligomeric complex. This was tested with the sequential use of BRET and FRET assays.209 BiFC assay was combined with BRET to show that the three receptors were in close proximity to interact.129 Although this finding is exciting, further studies are needed to determine whether these heterotrimers are present endogenously, and if so, what is their tissue distribution and physiological role.
2.4. Dopamine receptor heteromerization
Several studies have examined heteromerization between dopamine receptor subtypes and between the latter and other GPCRs. Since heteromerization between dopamine receptors and opioid receptor subtypes or CB1R have been described in Sections 2.2 and 2.3, we describe dopamine receptor heteromers that do not involve opioid or cannabinoid receptors in this section (see Table 9.3).
Table 9.3.
Heteromer pair | Detection techniques | Changes in heteromer properties | References | ||
---|---|---|---|---|---|
Binding | Signaling | Trafficking | |||
D1R–D2R | Co-IP, FRET, protein complementation assay | Not changed | Gαq-mediated signaling by the heteromer | Not reported | 125,220–224 |
D1R–D3R | Co-IP, BRET | ↑ Dopamine binding | ↑ Dopamine signaling | Heteromer internalized only by agonists to both receptors | 225 |
D1R–A1AR | Co-IP | Not reported | ↓ D1R signaling by combination of agonists to both receptors | Not reported | 226 |
D2R–D3R | Co-IP, functional complementation | Not reported | ↑ Potency with preferential protomer agonists | Not reported | 227,228 |
D2R–A2AR | Co-IP, BRET, FRET | Not reported | D2R ↓ efficacy and potency of A2AR agonists | Heteromer internalization by agonist to either protomer | 219,229–232 |
D2R–GSHR1a | TR-FRET | Not reported | D2R signals via Gαq and not Gαi | Not reported | 233 |
D2R–NTS1 | IP+fluorescence spectroscopy | ↓ Affinity for D2R agonist in the presence of NTS1 agonist | ↑ Potency of D2R in the presence of NTS1 agonist | NTS1 agonist ↑ D2R internalization | 234,235 |
D2R–OTR | BRET, proximity ligation assays | ↑ D2R antagonist binding by low doses of OTR agonist | ↑ Efficacy and potency of D2R signaling by OTR agonist | Not reported | 236 |
D2R–SSTR2 | Co-IP, pbFRET | Modulation of binding properties of individual protomers | SSTR2 agonist ↑ D2R signaling | D2R agonist ↑ SSTR2 internalization | 237 |
D2R–SSTR5 | Functional complementation, pbFRET | ↑ Affinity for each protomer agonist | Agonist to one protomer ↑ signaling by agonist to second protomer | Not reported | 144 |
D2R–5-HT2AR | Co-IP | D2R agonist ↑ affinity of 5-HT2AR agonist | D2R agonist ↑ potency of hallucinogenic 5-HT2AR agonist | Not reported | 238 |
D2R–mGluR5–A2AR | Co-IP, BRET+bimolecular fluorescence complementation | Not reported | Not reported | Not reported | 127 |
D4R–α1BAR | Co-IP, BRET | D4R agonist ↓ binding displacement by α1BAR agonist | D4R agonist ↓ α1BAR signaling | Not reported | 239 |
D4R–β1AR | Co-IP, BRET | D4R agonist ↓ binding displacement by β1AR agonist | D4R agonist ↓ β1AR signaling | Not reported | 239 |
D4R–β2AR | Co-IP, BRET | Not reported | Not reported | Not reported | 240 |
This table does not include heteromers involving opioid or cannabinoid receptors. These are described in Tables 9.1 and 9.2.
D1R–D2R heteromers
Interactions between D1R and D2R were suggested by the colocalization of D1R and D2R in MSNs in the striatum (reviewed in Refs. 48–50). Further evidence for direct interaction was suggested by the findings that (i) there is a requirement of D1R activation for D2R-mediated locomotor activity241; (ii) enhancement of the acute effects of cocaine requires activation of D1R and of D2R242; and (iii) potentiation in the release of arachidonic acid is observed upon coactivation of D1R and D2R in cells expressing both receptors.243
Evidence for direct interactions between D1R and D2R came from coimmunoprecipitation and confocal FRET studies carried out with rat striatal membranes, primary rat striatal neuronal cultures or with cells coexpressing epitope-tagged receptors.220–224 In addition, protein complementation studies where coexpression of D1R fused to a fragment of luciferase and of D2R fused to a complementary luciferase fragment resulted in the reconstitution of active luciferase supported D1R–D2R heteromerization.125 Signaling assays showed that while activation of D1R led to Gαs/olf-mediated signaling and of D2R to Gαi-mediated signaling, coactivation of both receptors led to increases in intracellular Ca2+ levels.220,221 Since the increase in intracellular Ca2+ levels is not observed in cells that express only D1R or D2R this suggested that the D1R–D2R heteromer can couple to Gαq to induce Ca2+-mediated signaling.220,221 Interestingly, occupancy of D1R in cells expressing D1R–D2R heteromers results in rapid desensitization of heteromer-mediated Ca2+ signaling that is regulated by both the catalytic and RGS domains of GRK2.223
The D1R agonist, SKF83959, has been proposed as a D1R–D2R heteromer-selective ligand based on the finding that (i) it did not increase adenylyl cyclase activity in cells coexpressing D1R–D2R or expressing only D1R; (ii) it increased intracellular Ca2+ release in cells coexpressing D1R–D2R, and this effect was potentiated by coadministration of quinpirole; and (iii) it increased [35S]GTPγS binding to Gαq and to Gαi in cells coexpressing D1R–D2R.221 Finally, D1R–D2R-mediated signaling by SKF83959 in primary striatal neurons via Gαq was found to lead to activation of both cytosolic and nuclear Ca2+/calmodulin-dependent kinase IIα resulting in increased expression of BDNF and ultimately rapid neuronal maturation and differentiation.222 Moreover, treatment with SKF83959 increased grooming behavior in rats and decreased the phosphorylation of the GluR1 subunit of AMPA receptors via Ca2+/calmodulin-dependent kinase IIα in the nucleus accumbens.224
D1R–D3R heteromers
Interactions between D1R and D3R were suggested by studies showing that (i) D1R agonists could induce the expression of mRNA and increase D3R levels, and that this could be blocked by a D1R antagonist244,245; (ii) the coactivation of D1R and D3R caused a potentiation in the expression of substance P mRNA246,247; (iii) D3R knockout mice exhibit a decrease in D1R-mediated expression of the immediate early gene, c-fos248; and (iv) cross talk between D1R and D3R may play a role in rearing habituation and reward behavior to low doses of cocaine.249
Evidence for direct interactions came from coimmunoprecipitation studies showing that D1R and D3R form interacting complexes in cells coexpressing both receptors and in rat striatal membranes.225 BRET assays showed that the two receptors were in close proximity in living cells.225 Treatment with dopamine increased the BRET signal, and this effect could be blocked by expression of a dominant negative mutant of dynamin leading to the suggestion that the increased signal could be due to internalized D1R–D3R heteromers.225 Binding studies with a radiolabeled D1R antagonist showed that dopamine binds with higher affinity to D1R in the presence of D3R.225 This is translated into more potent dopamine-mediated stimulation of adenylyl cyclase activity in cells coexpressing D1R and D3R than in cells expressing only D1R.225 Examination of the trafficking properties of D1R–D3R heteromer showed that treatment with either a D1R or a D3R agonist did not induce heteromer internalization; however, cotreatment with agonists to both receptors induced the internalization of the D1R–D3R heteromer.225
D2R–D3R heteromers
Functional complementation studies suggested that D2R and D3R could form heterodimers.227 For example, expression of D2R with a mutant D3R receptor (that cannot signal) restores the ability of D3R agonists to inhibit adenylyl cyclase activity.227 Coimmunoprecipitation assays with differentially epitope-tagged receptors showed that D2R and D3R could form interacting complexes.227 Signaling assays show that preferential D3R versus D2R agonists (pramipexole, ropinirole, or S32504) were more potent at inhibiting the activity of a chimeric adenylyl cyclase (does not respond to D3R activation) in cells coexpressing D2R and D3R compared to cells expressing only D2R (reviewed in Ref. 228). In contrast, preferential D2R agonists such as pergolide showed a marginal increase in potency in cells coexpressing the D2R–D3R heteromer while no changes were observed for the full D2R/D3R agonist, quinpirole (reviewed in Ref. 228). Interestingly, partial D2R/D3R agonists with antipsychotic activity did not inhibit adenylyl cyclase activity in cells coexpressing both receptors and also could block quinpirole-mediated inhibition of cAMP levels (reviewed in Ref. 228). This suggests that in cells expressing D2R–D3R heteromers partial D2R/D3R agonists can function as antagonists (reviewed in Ref. 228).
D1R–adenosine A1AR heteromers
Interactions between D1R and A1AR receptors were suggested by studies showing that (i) A1AR activation leads to a decrease in D1R-mediated locomotion and oral dyskinesia in animals,250 while A1AR antagonists enhance D1R-mediated locomotion251; (ii) radiolabeled D1R antagonist affinity is decreased by A1AR agonist252; (iii) A1AR agonists modulate D1R-mediated electroencephalographic arousal11; and (iv) in cells coexpressing A1AR and D1R, A1AR antagonists cause an increase in both basal- and dopamine-mediated increases in cAMP levels while an A1AR agonist blocks dopamine-mediated effects.253
Coimmunoprecipitation studies using receptor-selective antibodies detected the presence of D1R–A1AR interacting complexes in cells coexpressing both receptors.226 In fibroblast cells that coexpress D1R and A1AR, pretreatment with agonists to both receptors decreases D1R-mediated increases in cAMP levels, while pretreatment with either receptor agonist does not. Additional studies using proximity-based assays to confirm the formation of D1R–A1AR heteromers in vivo as well as studies to evaluate whether the D1R–A1AR heteromer exhibits novel properties compared to individual receptor homomers need to be carried out.
D2R–adenosine A2AR heteromers
Interactions between D2R and A2AR are suggested by studies showing that (i) D2R and A2AR are colocalized in GABAergic striatopallidal neurons254; (ii) selective A2AR agonists decrease D2R binding and signaling229,255,256; (iii) Blockade or knockdown of D2R impairs locomotive behavior, and this effect can be attenuated by A2AR antagonists257,258; and (iv) antagonists of D2R produce catalepsy and this is blocked by A2AR antagonists.259 The formation of interacting D2R–A2AR complexes was demonstrated by coimmunoprecipitation studies and by proximity-based assays such as BRET and FRET.219,230–232 Investigation of the pharmacological properties of the D2R–A2AR in cells coexpressing both receptors showed that D2R agonists caused a decrease in the affinity for radiolabeled A2AR agonist binding.229 Moreover, signaling assays showed that D2R agonists decreased the efficacy and potency of A2AR agonists in cells coexpressing both receptors.229 In addition, studies showed that treatment with either A2AR or the D2R agonists causes internalization of the D2R–A2AR heteromer.219,260
Bivalent ligands comprising of a D2R agonist joined by spacers of different lengths (26–118 atoms) to an A2AR antagonist have been used to show that in striatal membranes coexpressing both receptors, these ligands cause a greater displacement of radiolabeled ligand binding to either D2R or A2AR than the corresponding monovalent ligands.261
D2R–ghrelin receptor heteromers
A recent study examined D2R and ghrelin receptor (GHSR1a) interactions because of their robust colocalization to a subset of hypothalamic neurons.233 Time-resolved FRET carried out in heterologous cells and with hypothalamic membrane preparations demonstrated that the two receptors are in close proximity to form heteromers.233 In cells coexpressing D2R and GHSR1a, activation of D2R led to increases in intracellular Ca2+ release and not to Gαi-mediated signaling and this could be blocked by a D2R or a GHSR1a antagonist.233 Moreover, cotreatment with agonists to both receptors led to additive effects on intracellular Ca2+ release.233 In addition, cross-desensitization of Ca2+ release is observed in cells coexpressing both receptors in that a 30-min pretreatment with agonist to one receptor attenuated intracellular Ca2+ release mediated via the other receptor.233 Studies with wild-type and ghrelin receptor knockout mice showed that administration of a D2R agonist caused a reduction in food intake in wild-type but not in knockout mice.233 This reduction in food intake can be blocked by a GHSR1a antagonist.233 These results indicate that D2R–GHSR1a heteromerization could play an important role in the modulation of food intake.
D2R–neurotensin receptor heteromers
Interactions between dopamine and neurotensin receptors were demonstrated by studies showing that (i) type 1 neurotensin receptors (NTS1) are expressed in substantia nigra and ventral tegmental area dopaminergic neurons262; (ii) NTS1 has excitatory effects on dopaminergic neurons present in the midbrain262; (iii) neurotensin causes a modest decrease in the affinity of D2R for receptor agonists262; (iv) D2R autoreceptor function is decreased by NTS1 activation via a protein kinase C- and Ca2+-dependent mechanism262; and (v) activation of D2R attenuates NTS1-mediated Ca2+ mobilization.262
A combination of immunoprecipitation of the long isoform of D2R (D2RL) with fluorescent spectroscopy for detection of the presence of C-terminally epitope tagged (with enhanced CFP) NTS1 in the immunoprecipitate indicated that the two receptors formed interacting complexes only in cells coexpressing both receptors.234 Radiolabeled binding experiments with membrane preparations from cells coexpressing D2RL and NTS1 showed that the NTS1 agonist decreased the affinity of radiolabeled agonist binding to D2RL.234 However, dopamine ligands (agonist or antagonist) had no effect on radiolabeled agonist binding to NTS1.234 Signaling assays showed that the potency of D2RL-mediated signaling is increased in the presence of neurotensin.234 Internalization assays showed that in cells coexpressing D2RL and NTS1 treatment with NTS1 agonist increased D2RL internalization via a PKC-dependent mechanism.235
D2R–oxytocin receptor heteromers
Studies showing (i) the involvement of striatal oxytocin receptors (OTR) in pair bond formation in monogamous prairie female voles and in regulating social and emotional behaviors in animals (rodents, sheep, humans)263–267; (ii) the involvement of ascending dopaminergic neurons in social attachment behavior1,2; (iii) that a D2R antagonist can block partner preference268,269; and (iv) administration of a D2R agonist into the nucleus accumbens can induce partner preference and this is blocked by an OTR antagonist268,269 suggest interactions between D2R and OTR.
BRET assays in cells coexpressing both receptors showed that D2R and OTR are in close proximity in live cells.236 In situ proximity ligation assays detected the presence of D2R–OTR heteromers in the neuropil of nucleus accumbens and in the dorsal striatum of rats.236 Binding assays with nucleus accumbens membrane preparations showed that a low, but not high, concentration of the OTR agonist increase the maximal number of binding sites of a radiolabeled D2R antagonist and this is blocked by an OTR antagonist.236 Signaling assays with nucleus accumbens membrane preparations showed that the OTR agonist increases the efficacy and potency of D2R-mediated G-protein activity.236 These studies indicate that D2R–OTR heteromers may play a role in modulating social and emotional behavior.
D2R–somatostatin receptor heteromers
Interactions between dopamine and somatostatin receptors were suggested by studies showing that (i) a decrease of somatostatin levels attenuated dopamine receptor-mediated stereotypy as well as locomotor responses to amphetamine270; (ii) dopamine enhances somatostatin receptor-mediated inhibition of adenylyl cyclase activity271,272; and that (iii) a dopamine receptor agonist attenuates the chronic effects of a long-acting somatostatin analogue.273 This led investigators to examine heteromerization between D2R and somatostatin receptor subtypes.
Direct interactions between D2R and somatostatin receptor 2 subtype (SSTR2) was examined by coimmunoprecipitation and pbFRET assays that demonstrated that these two receptors formed complexes in heterologous cells coexpressing both receptors as well as in primary striatal neuronal cultures.237 Binding studies showed that in cells coexpressing D2R and SSTR2 a combination of agonists to both receptors displaced radiolabeled antagonist binding to D2R more efficiently than the D2R agonist alone.237 Interestingly, in these cells, the SSTR2 agonist could dose-dependently displace radiolabeled antagonist binding to D2R.237 Also, the total number of binding sites for the radiolabeled ligand binding to SSTR2 was increased in the presence of dopamine.237 Examination of signaling properties of the D2R–SSTR2 heteromer showed that D2R-mediated inhibition of cAMP levels was lower for some agonists (dopamine) and not others (quinpirole) than that observed in cells that expressed only D2R.237 Moreover, the presence of the SSTR2 agonist caused an increase in the potency of quinpirole and in both the potency and efficacy of dopamine in inhibiting cAMP levels.237 Examination of the trafficking properties of the D2R–SSTR2 heteromer showed that treatment with D2R agonist enhanced agonist-mediated internalization of SSTR2.237
Heteromerization between D2R and SSTR5 was suggested by the functional rescue of a C-tail deletion mutant of human SSTR5.144 Only when coexpressed with D2R does agonist activation of the mutant SSTR5 lead to inhibition in forskolin-mediated increases in cAMP levels.144 pbFRET supported the formation of D2R–SSTR5 heteromers.144 Interestingly, treatment with agonists to either receptor or cotreatment with agonists to both receptors led to an increase in FRET efficiency, while a decrease in the latter was observed with D2R antagonist.144 This would suggest that either agonist treatment leads to an increase in the level of D2R–SSTR5 heteromers or, alternatively, it changes receptor conformation leading to a decrease in the distance between the fluorescent probes.144 The pharmacological properties of the D2R–SSTR5 heteromer were found to be distinct from those of individual receptor homomers in that the heteromers exhibited a greater affinity for the binding of each protomer agonist.144 Moreover, occupancy of one of the receptors in the D2R–SSTR5 heteromer led to an increase in the binding affinity and signaling by the agonist to the second receptor.144
D2R–serotonin receptor heteromers
Studies showing that typical antipsychotic drugs used to treat schizophrenia are D2R antagonists and that some symptoms of schizophrenia are similar to those observed with hallucinogenic serotonin receptor agonists led to the examination of heteromerization between D2R and serotonin 5-HT2A receptors (5-HT2AR).274,275
Coimmunoprecipitation studies show that D2R and 5-HT2AR form interacting complexes.238 Examination of the pharmacological properties of the D2R–5-HT2AR heteromer in cells or in striatal membranes coexpressing both receptors show that a D2R agonist increased the binding affinity of a 5-HT2AR agonist, and this is blocked by a D2R antagonist.238 The presence of D2R had no effect on serotonin-mediated inositol phosphate production, but it increased the potency of a hallucinogenic 5-HT2AR agonist.238 Interestingly, D2R activation had no effect on serotonin-mediated inositol phosphate production, but decreased the potency of the hallucinogenic 5-HT2AR agonist.238 Behavioral studies with 5-HT2AR knockout mice show that the antipsychotic effects of the D2R antagonist, haloperidol, require the presence of 5-HT2AR.238
D4R–α adrenergic receptor heteromers
Heteromer formation between D4R and α adrenergic receptors (αAR) was examined based on studies showing that the D4R gene is the most prominently expressed dopamine receptor gene in the rat pineal gland, and that its mRNA expression is circadian and controlled by adrenergic signaling.276
Coimmunoprecipitation and BRET studies with differentially epitope-tagged receptors show that D4R and α1BAR form interacting complexes.239 Pharmacological characterization of D4R–adrenergic receptor heteromers shows that the D4R agonist decreases the ability of an α1BAR agonist to displace radiolabeled antagonist binding to α1BAR.239 Signaling assays in cells expressing the D4R–α1BAR heteromer or in pineal gland preparations show that pretreatment or cotreatment with a selective D4R agonist leads to an inhibition of α1BAR-mediated phosphorylation of ERK1/2 and of Akt/PKB.239 Interestingly, a D4R receptor antagonist could also block α1BAR-mediated signaling and the converse also occurred in that adrenergic antagonists could block D4R-mediated signaling.239 Given that D4R expression is circadian, and that α1BAR plays a role in modulating melatonin synthesis and release from the pineal gland, the formation of D4R–α1BAR heteromers provides a means for regulating melatonin production and activity.239
D4R–β1 adrenergic receptor heteromers
Studies examining D4R–αAR heteromerization also examined whether D4R and β1 adrenergic receptors (β1AR) formed heteromers. Coimmunoprecipitation and BRET studies with differentially epitope-tagged receptors show that D4R and β1AR form interacting complexes.239 Pharmacological characterization of D4R–β1AR heteromers shows that the D4R agonist decreases the ability of a β1AR agonist to displace radiolabeled antagonist binding to β1AR.239 Signaling assays in cells expressing either the D4R–β1AR heteromer or in pineal gland preparations show that pretreatment or cotreatment with a selective D4R agonist leads to an inhibition of β1AR-mediated phosphorylation of ERK1/2 and of Akt/PKB.239 Interestingly, a D4R receptor antagonist could also block β1AR-mediated signaling and the converse also occurred in that adrenergic antagonists could block D4R-mediated signaling.239 These studies suggest that like the D4R–α1BAR heteromer the D4R–β1AR heteromer could also play a role in regulating melatonin production and activity.239
D4R–β2 adrenergic receptor heteromers
Based on studies showing (i) colocalization of D2R-like receptors with β-AR in rat primary cortical neurons and in prefrontal cortex277 and that (ii) D2R-like receptor activation inhibited β-AR-mediated increases in cAMP levels,277 heteromerization between D4R and β2-AR was examined.
Coimmunoprecipitation, BRET, and binding assays with β2-AR immunoprecipitates, obtained from detergent solubilized mouse brain extracts using alprenolol coupled to Sepharose beads, show that D4R and β2-AR form interacting complexes and are in close proximity in cells or tissues coexpressing both receptors.240 However, further studies are needed to evaluate whether the D4R–β2-AR heteromer exhibits novel properties compared to individual receptor homomers.
D2R–metabotropic glutamate receptor 5–adenosine2A receptor heteromers
The possibility of formation of heteromers involving D2R, metabotropic glutamate receptor 5 (mGluR5), and adenosine2A receptors (A2AR) was suggested by studies showing (i) similar localization of these receptors in striatopallidal neurons278–280; (ii) that a mGluR5 agonist inhibited D2R-mediated contralateral turning behavior, and this was potentiated by an A2AR agonist and attenuated by an A2AR antagonist281; and (iii) that a mGluR5 agonist reduced the high affinity state for the D2R agonist, and this was potentiated by an A2AR agonist.281
Coimmunoprecipitation studies carried out with cells coexpressing D2R, A2AR, and mGluR5, and also with rat striatal membranes, show that these receptors form interacting complexes.127 BRET assays with cells expressing a constant ratio of A2AR tagged with luciferase and D2R tagged with YFP show that increasing amounts of mGluR5 can disrupt the BRET signal.127 Similarly, increasing amounts of A2AR could disrupt the BRET signal between D2R tagged with luciferase and mGluR5 tagged with YFP.127 This suggests that the three receptors are in close proximity to interact.127 The formation of D2R–A2AR–mGluR5 was confirmed by a combination of BRET and BiFC assays.127 For this, cells were transfected with A2AR tagged with luciferase, mGluR5 fused with an N-terminal fragment of YFP and D2R fused with a C-terminal fragment of YFP.127 A BRET signal was observed only when all three receptors were expressed in a cell suggesting that these receptors are in close proximity to form heterotrimers.127 Further studies are needed in order to elucidate whether the D2R–A2AR–mGluR5 heterotrimer exhibits novel properties compared to individual receptor homomers or heterodimers.
3. HETEROMERS IN DISEASE
Although it has become increasingly evident that GPCRs can form heterodimers,61,63,64,282 very littleinformation is available aboutthe physiological role of GPCR heteromers. Pharmacological and coimmunoprecipitation studies using receptor-selective antibodies suggest that some receptor heteromers may play a role in the disease state. However, care has to be taken in the interpretation of such data given that such studies do not rule out a contribution of receptor homomers to the disease state. The recent development of antibodies that selectively recognize receptor heteromers, but not receptor homomers, and of TAT-based peptides that can disrupt heteromer formation, has provided us with tools to study the role of GPCR heteromers in the disease state.7–10 In the following sections, we describe a few cases where a role for GPCR heteromers involving opioid, cannabinoid, and dopamine receptors in pathology has been implicated.
Analgesia and addiction
Antibodies that selectively recognize the MOR–DOR heteromer detect increased heteromer levels in brain regions involved in pain processing following chronic morphine administration under a paradigm that leads to the development of tolerance.7 Taken with the fact that MOR–DOR heteromers signal via the β-arrestin-mediated signaling cascade100 and that β-arrestin or DOR knockout mice do not develop tolerance to morphine162,283 would suggest that MOR–DOR heteromers play a role in the development of tolerance to this opioid. This is supported by a study showing that the administration of a MOR transmembrane 1 TAT peptide fusion protein, which selectively disrupts the MOR–DOR heteromer, resulted in an enhanced analgesic response to morphine with a concomitant decrease in tolerance to the drug.8
Studies have also examined whether the DOR–MOR heteromer could be a _target for the development of therapeutics that are as efficacious as morphine but lack the associated adverse side effects. In this context, a study showed that low doses of the DOR antagonists, TIPPψ or naltrindole, could potentiate intrathecal morphine-mediated analgesia.94,160 Moreover, DOR antagonist administration to animals exhibiting tolerance to morphine led to a restoration of the analgesic response and potency of morphine.160 Based on similar observations, bivalent ligands _targeting the DOR–MOR heteromer were developed.284 These bivalent ligands comprised a MOR agonist pharmacophore, oxymorphone, joined to a DOR antagonist pharmacophore, naltrindole, by spacers of variable lengths (16–21 atoms).284 The bivalent compounds exhibited antinociceptive activity.284 Moreover, compounds with spacer lengths between 19 and 21 atoms did not exhibit antinociceptive tolerance and physical dependence and were less rewarding than morphine.284,285
An involvement of DOR–KOR heteromers in thermal allodynia was suggested by studies demonstrating the presence of these heteromers in peripheral sensory neurons.286 Moreover, the antiallodynic effects of the KOR agonist, 6′-GNTI, to prostaglandin E2-induced thermal allodynia were blocked by either DOR or KOR antagonists.286 This, together with the finding that the antiallodynic effects of the DOR agonist, DPDPE, were potentiated by a DOR–KOR heteromer-selective antibody supports a role for the DOR–KOR heteromer in modulating thermal allodynia.286
A recent study has implicated the CB1R–DOR heteromer in neuropathic pain.9 This study found that levels of CB1R and DOR are increased in distinct brain regions following peripheral nerve lesion.9 An antibody selective for the CB1R–DOR heteromer detected increased heteromer levels in the cortex, hypothalamus, and striatum of lesioned animals.9 In addition, CB1R–DOR heteromerization led to a decrease in DOR activity in the cortex of lesioned animals9 and this decrease could be attenuated by low, nonsignaling doses of CB1R ligands.9 Thus, the CB1R–DOR heteromer is a novel _target for the treatment of neuropathic pain.
Liver fibrosis
Antibodies that selectively recognize the CB1R–AT1R heteromer detect a significant upregulation of this heteromer in hepatic stellate cells of rats chronically treated with ethanol.10 This leads to increased AT1R-mediated signaling that can be blocked by a CB1R antagonist or by inhibiting the synthesis of the endocannabinoid, 2-AG.10 In addition, the AT1R-mediated increase in profibrogenic markers in hepatic stellate cells of rats chronically treated with ethanol was completely blocked by a CB1R antagonist.10 These results suggest that the profibrogenic activity of AT1R in ethanol-induced liver fibrosis requires CB1R activity. Thus, the CB1R–AT1R heteromer represents a novel therapeutic _target for the treatment of this disease.
Psychiatric disorders
An involvement of dopamine receptor subtype heteromers in major depression has been suggested by studies showing an increase in D1R–D2R interacting complexes in the striatum of postmortem individuals that suffered from major depression.287 Interestingly, bilateral infusion into the rat prefrontal cortex of a TAT peptide that disrupts D1R–D2R heteromers results in a decrease in the levels of D1R–D2R heteromers and leads to antidepressant-like effects in the forced swim test.287 This suggests an involvement of D1–D2 receptor heteromers in the pathology associated with major depression.
Involvement of D1R–D2R heteromers in schizophrenia was suggested by studies showing that clozapine, a drug used to treat this condition, binds to both D1R and D2R but that the high affinity binding to D1R decreases in the presence of D2R.288,289 Moreover, FRET assays showed that clozapine appears to modulate the formation of D1R–D2R heteromers in that high concentrations of the drug increase the FRET signal between D1R and D2R while low concentrations attenuate this signal.288,289 Further support for the involvement D1R–D2R heteromers in schizophrenia came from studies showing that acute treatment with a heteromer-selective agonist increased the striatal expression of brain-derived neurotrophic factor, a molecule that has been linked to schizophrenia (for review, see Ref. 290).
An involvement of D2R–5-HT2AR heteromers in schizophrenia is suggested by studies showing that coexpression of D2R in cells increases the signaling potency of a hallucinogenic 5-HT2AR agonist, but activation of D2R decreases it.238 Moreover, the antipsychotic effects of the D2R antagonist, haloperidol, are decreased in 5-HT2AR knockout mice238 suggesting that the D2R–5-HT2AR heteromer is a _target of this drug.
An involvement of CB1R–D2R–A2AR heteromers in schizophrenia has been suggested based on studies showing upregulation of CB1R, D2R, and A2AR in the postmortem brains of schizophrenics.291–293 Moreover, in animal models of schizophrenia, treatment with antipsychotics decreases CB1R levels in the nucleus accumbens.294 These reports together with studies describing CB1R–D2R and CB1R–A2AR interactions (described in Section 2) and studies showing that these three receptors can form heteromers in live cells suggest that the CB1R–D2R–A2AR heteromer could be a novel therapeutic _target in the treatment of schizophrenia.
Parkinson’s disease
The involvement of D2R–A2AR heteromers in Parkinson’s disease is suggested by studies showing that the antagonistic D2R–A2AR interactions were increased in the striatal membranes from rat models of Parkinson’s disease (reviewed in Ref. 295). In addition, studies indicate that A2AR antagonists have anti-Parkinsonian effects (reviewed in Ref. 295). For example, A2AR antagonist administration could attenuate dopamine-mediated neuronal degeneration in animal models of Parkinson’s disease (reviewed in Ref. 295). Given that D2R–A2AR heteromers are constitutively formed, it is possible that A2AR antagonists _targeting the heteromer could increase D2R-mediated signaling leading, in turn, to an attenuation of Parkinson’s disease symptoms (reviewed in Ref. 295).
The heterotrimer involving D2R–mGluR5–A2AR could also be a novel therapeutic _target in the treatment of Parkinson’s disease (reviewed in Ref. 295). This is supported by studies showing that mGluR5 antagonist-mediated induction of motor activity requires the presence of both D2R and A2AR and that a combination of mGluR5 and A2AR antagonists could potentiate locomotor behavior in reserpinized mice (reviewed in Ref. 295).
Another heterotrimer that could be a novel _target for the treatment of Parkinson’s disease involves CB1R–D2R–A2AR (reviewed in Ref. 295). Low doses of CB1R and A2AR antagonists could synergize to potentiate D2R-mediated anti-Parkinsonian effects during the early stages of Parkinson’s disease when a substantial release of dopamine still occurs (reviewed in Ref. 295).
4. CONCLUSIONS
Recent evidence indicates that some GPCR heteromers including those associated with drugs of abuse are upregulated under pathophysiological conditions. However, the physiological role of these heteromers in pathology is not clearly understood due to the lack of tools to distinguish between heteromer and homomer-mediated effects. For this, heteromer-specific ligands, agonists and/or antagonists, are needed to elucidate the role of GPCR heteromers in normal and pathophysiological conditions. Although heteromer-specific ligands for a few GPCR heteromers have been reported they are mostly receptor agonists and need to be extensively evaluated for their heteromer selectivity in both in vitro and in vivo assays. Moreover, the availability of heteromer-selective antagonists would help in the evaluation of heteromer levels in the normal and diseased states as well as in studies examining heteromer pharmacology. The recent development of antibodies that selectively recognize a GPCR heteromer in endogenous tissue, and of TAT peptides that can specifically disrupt the heteromer of interest, could help to elucidate the role of GPCR heteromers in pathological conditions.7–10 This is clearly shown in the case of MOR–DOR heteromers where heteromer-selective antibodies demonstrate that these heteromers are increased in brain areas involved in pain perception following chronic treatment with morphine under a paradigm that leads to the development of tolerance.7 In addition, studies with TAT peptides that selectively disrupt the MOR–DOR heteromer suggest a role for the heteromer to keep morphine-mediated signaling via MOR in the desensitized state.8
Studies demonstrating that GPCR heteromers are upregulated during a disease state suggest that these heteromers could be novel _targets for the development of therapeutics to treat these diseases. If heteromers are disease-specific and/or exhibit unique tissue specificity, they would serve as ideal drugs for the development of therapeutics with potentially lesser side effects than the ones that are currently available.
Acknowledgments
We thank Jonathan Wardman and Steven D. Stockton, Jr. for critical reading and helpful comments. L. A. D. is supported by NIH grants DA008863 and DA019521.
References
- 1.Kieffer BL. Recent advances in molecular recognition and signal transduction of active peptides: receptors for opioid peptides. Cell Mol Neurobiol. 1995;15:615–35. doi: 10.1007/BF02071128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Kieffer BL. Opioids: first lessons from knockout mice. Trends Pharmacol Sci. 1999;20:19–26. doi: 10.1016/s0165-6147(98)01279-6. [DOI] [PubMed] [Google Scholar]
- 3.Howlett AC, Barth F, Bonner TI, Cabral G, Casellas P, Devane WA, et al. International Union of Pharmacology. XXVII. Classification of cannabinoid receptors. Pharmacol Rev. 2002;54:161–202. doi: 10.1124/pr.54.2.161. [DOI] [PubMed] [Google Scholar]
- 4.Cichewicz DL. Synergistic interactions between cannabinoid and opioid analgesics. Life Sci. 2004;74:1317–24. doi: 10.1016/j.lfs.2003.09.038. [DOI] [PubMed] [Google Scholar]
- 5.Vigano D, Rubino T, Parolaro D. Molecular and cellular basis of cannabinoid and opioid interactions. Pharmacol Biochem Behav. 2005;81:360–8. doi: 10.1016/j.pbb.2005.01.021. [DOI] [PubMed] [Google Scholar]
- 6.Sulzer D. How addictive drugs disrupt presynaptic dopamine neurotransmission. Neuron. 2011;69:628–49. doi: 10.1016/j.neuron.2011.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Gupta A, Mulder J, Gomes I, Rozenfeld R, Bushlin I, Ong E, et al. Increased abundance of opioid receptor heteromers after chronic morphine administration. Sci Signal. 2010;3:ra54. doi: 10.1126/scisignal.2000807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.He SQ, Zhang ZN, Guan JS, Liu HR, Zhao B, Wang HB, et al. Facilitation of mu-opioid receptor activity by preventing delta-opioid receptor-mediated code-gradation. Neuron. 2011;69:120–31. doi: 10.1016/j.neuron.2010.12.001. [DOI] [PubMed] [Google Scholar]
- 9.Bushlin I, Gupta A, Stockton SD, Jr, Miller LK, Devi LA. Dimerization with cannabinoid receptors allosterically modulates delta opioid receptor activity during neuropathic pain. PLoS One. 2012;7:e49789. doi: 10.1371/journal.pone.0049789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Rozenfeld R, Gupta A, Gagnidze K, Lim MP, Gomes I, Lee-Ramos D, et al. AT1R-CBR heteromerization reveals a new mechanism for the pathogenic properties of angiotensin II. EMBO J. 2011;30:2350–63. doi: 10.1038/emboj.2011.139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Popoli P, Ferre S, Pezzola A, Reggio R, deCarolis AS, Fuxe K. Stimulation of adenosine A(1) receptors prevents the EEG arousal due to dopamine D-1 receptor activation in rabbits. Eur J Pharmacol. 1996;305:123–6. doi: 10.1016/0014-2999(96)00242-7. [DOI] [PubMed] [Google Scholar]
- 12.Berezniuk I, Fricker LD. Endogenous opioids. In: Pasternak GW, editor. The opiate receptors. Vol. 23. New York: Humana Press; 2011. pp. 93–120. [Google Scholar]
- 13.Akil H, Watson SJ, Young E, Lewis ME, Khachaturian H, Walker JM. Endogenous opioids: biology and function. Annu Rev Neurosci. 1984;7:223–55. doi: 10.1146/annurev.ne.07.030184.001255. [DOI] [PubMed] [Google Scholar]
- 14.Suh HH, Tseng LF. Delta but not mu-opioid receptors in the spinal cord are involved in antinociception induced by beta-endorphin given intracerebroventricularly in mice. J Pharmacol Exp Ther. 1990;253:981–6. [PubMed] [Google Scholar]
- 15.Sim LJ, Selley DE, Dworkin SI, Childers SR. Effects of chronic morphine administration on mu opioid receptor-stimulated [35S]GTPgammaS autoradiography in rat brain. J Neurosci. 1996;16:2684–92. doi: 10.1523/JNEUROSCI.16-08-02684.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Matthes HW, Maldonado R, Simonin F, Valverde O, Slowe S, Kitchen I, et al. Loss of morphine-induced analgesia, reward effect and withdrawal symptoms in mice lacking the mu-opioid-receptor gene. Nature. 1996;383:819–23. doi: 10.1038/383819a0. [DOI] [PubMed] [Google Scholar]
- 17.Ghozland S, Matthes HW, Simonin F, Filliol D, Kieffer BL, Maldonado R. Motivational effects of cannabinoids are mediated by mu-opioid and kappa-opioid receptors. J Neurosci. 2002;22:1146–54. doi: 10.1523/JNEUROSCI.22-03-01146.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Roberts AJ, McDonald JS, Heyser CJ, Kieffer BL, Matthes HW, Koob GF, et al. mu-Opioid receptor knockout mice do not self-administer alcohol. J Pharmacol Exp Ther. 2000;293:1002–8. [PubMed] [Google Scholar]
- 19.Moles A, Kieffer BL, D’Amato FR. Deficit in attachment behavior in mice lacking the mu-opioid receptor gene. Science. 2004;304:1983–6. doi: 10.1126/science.1095943. [DOI] [PubMed] [Google Scholar]
- 20.Kieffer BL, Evans CJ. Opioid receptors: from binding sites to visible molecules in vivo. Neuropharmacology. 2009;56(Suppl 1):205–12. doi: 10.1016/j.neuropharm.2008.07.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Filliol D, Ghozland S, Chluba J, Martin M, Matthes HW, Simonin F, et al. Mice deficient for delta- and mu-opioid receptors exhibit opposing alterations of emotional responses. Nat Genet. 2000;25:195–200. doi: 10.1038/76061. [DOI] [PubMed] [Google Scholar]
- 22.Simonin F, Valverde O, Smadja C, Slowe S, Kitchen I, Dierich A, et al. Disruption of the kappa-opioid receptor gene in mice enhances sensitivity to chemical visceral pain, impairs pharmacological actions of the selective kappa-agonist U-50,488H and attenuates morphine withdrawal. EMBO J. 1998;17:886–97. doi: 10.1093/emboj/17.4.886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Land BB, Bruchas MR, Lemos JC, Xu M, Melief EJ, Chavkin C. The dysphoric component of stress is encoded by activation of the dynorphin kappa-opioid system. J Neurosci. 2008;28:407–14. doi: 10.1523/JNEUROSCI.4458-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Zhao P, Abood ME. GPR55 and GPR35 and their relationship to cannabinoid and lysophospholipid receptors. Life Sci. 2012;92:453–7. doi: 10.1016/j.lfs.2012.06.039. [DOI] [PubMed] [Google Scholar]
- 25.Di Marzo V, Petrosino S. Endocannabinoids and the regulation of their levels in health and disease. Curr Opin Lipidol. 2007;18:129–40. doi: 10.1097/MOL.0b013e32803dbdec. [DOI] [PubMed] [Google Scholar]
- 26.Battista N, Fezza F, Finazzi-Agro A, Maccarrone M. The endocannabinoid system in neurodegeneration. Ital J Biochem. 2006;55:283–9. [PubMed] [Google Scholar]
- 27.Boyd ST. The endocannabinoid system. Pharmacotherapy. 2006;26:218S–221S. doi: 10.1592/phco.26.12part2.218S. [DOI] [PubMed] [Google Scholar]
- 28.Okamoto Y, Morishita J, Tsuboi K, Tonai T, Ueda N. Molecular characterization of a phospholipase D generating anandamide and its congeners. J Biol Chem. 2004;279:5298–305. doi: 10.1074/jbc.M306642200. [DOI] [PubMed] [Google Scholar]
- 29.Simon GM, Cravatt BF. Endocannabinoid biosynthesis proceeding through glycerophospho-N-acyl ethanolamine and a role for alpha/beta-hydrolase 4 in this pathway. J Biol Chem. 2006;281:26465–72. doi: 10.1074/jbc.M604660200. [DOI] [PubMed] [Google Scholar]
- 30.Liu J, Wang L, Harvey-White J, Osei-Hyiaman D, Razdan R, Gong Q, et al. A biosynthetic pathway for anandamide. Proc Natl Acad Sci USA. 2006;103:13345–50. doi: 10.1073/pnas.0601832103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Prescott SM, Majerus PW. Characterization of 1,2-diacylglycerol hydrolysis in human platelets. Demonstration of an arachidonoyl-monoacylglycerol intermediate. J Biol Chem. 1983;258:764–9. [PubMed] [Google Scholar]
- 32.Farooqui AA, Taylor WA, Horrocks LA. Separation of bovine brain mono- and diacylglycerol lipases by heparin sepharose affinity chromatography. Biochem Biophys Res Commun. 1984;122:1241–6. doi: 10.1016/0006-291x(84)91225-7. [DOI] [PubMed] [Google Scholar]
- 33.Cravatt BF, Demarest K, Patricelli MP, Bracey MH, Giang DK, Martin BR, et al. Supersensitivity to anandamide and enhanced endogenous cannabinoid signaling in mice lacking fatty acid amide hydrolase. Proc Natl Acad Sci USA. 2001;98:9371–6. doi: 10.1073/pnas.161191698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Kathuria S, Gaetani S, Fegley D, Valino F, Duranti A, Tontini A, et al. Modulation of anxiety through blockade of anandamide hydrolysis. Nat Med. 2003;9:76–81. doi: 10.1038/nm803. [DOI] [PubMed] [Google Scholar]
- 35.Hampson AJ, Hill WA, Zan-Phillips M, Makriyannis A, Leung E, Eglen RM, et al. Anandamide hydroxylation by brain lipoxygenase:metabolite structures and potencies at the cannabinoid receptor. Biochim Biophys Acta. 1995;1259:173–9. doi: 10.1016/0005-2760(95)00157-8. [DOI] [PubMed] [Google Scholar]
- 36.Yu M, Ives D, Ramesha CS. Synthesis of prostaglandin E2 ethanolamide from anandamide by cyclooxygenase-2. J Biol Chem. 1997;272:21181–6. doi: 10.1074/jbc.272.34.21181. [DOI] [PubMed] [Google Scholar]
- 37.Dinh TP, Carpenter D, Leslie FM, Freund TF, Katona I, Sensi SL, et al. Brain monoglyceride lipase participating in endocannabinoid inactivation. Proc Natl Acad Sci USA. 2002;99:10819–24. doi: 10.1073/pnas.152334899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Gomes I, Grushko JS, Golebiewska U, Hoogendoorn S, Gupta A, Heimann AS, et al. Novel endogenous peptide agonists of cannabinoid receptors. FASEB J. 2009;23:3020–9. doi: 10.1096/fj.09-132142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Heimann AS, Gomes L, Dale CS, Pagano RL, Gupta A, de Souza LL, et al. Hemopressin is an inverse agonist of CB1 cannabinoid receptors. Proc Natl Acad Sci USA. 2007;104:20588–93. doi: 10.1073/pnas.0706980105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Gomes I, Dale CS, Casten K, Geigner MA, Gozzo FC, Ferro ES, et al. Hemoglobin-derived peptides as novel type of bioactive signaling molecules. AAPS J. 2010;12:658–69. doi: 10.1208/s12248-010-9217-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Gelman JS, Sironi J, Castro LM, Ferro ES, Fricker LD. Hemopressins and other hemoglobin-derived peptides in mouse brain: comparison between brain, blood, and heart peptidome and regulation in Cpefat/fat mice. J Neurochem. 2010;113:871–80. doi: 10.1111/j.1471-4159.2010.06653.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Gelman JS, Fricker LD. Hemopressin and other bioactive peptides from cytosolic proteins: are these non-classical neuropeptides? AAPS J. 2010;12:279–89. doi: 10.1208/s12248-010-9186-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Bushlin I, Rozenfeld R, Devi LA. Cannabinoid-opioid interactions during neuropathic pain and analgesia. Curr Opin Pharmacol. 2010;10:80–6. doi: 10.1016/j.coph.2009.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Miller LK, Devi LA. The highs and lows of cannabinoid receptor expression in disease: mechanisms and their therapeutic implications. Pharmacol Rev. 2011;63:461–70. doi: 10.1124/pr.110.003491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Valverde O, Torrens M. Cbi receptor-deficient mice as a model for depression. Neuroscience. 2012;204:193–206. doi: 10.1016/j.neuroscience.2011.09.031. [DOI] [PubMed] [Google Scholar]
- 46.Valverde O, Karsak M, Zimmer A. Analysis of the endocannabinoid system by using CB1 cannabinoid receptor knockout mice. Handb Exp Pharmacol. 2005;168:117–45. doi: 10.1007/3-540-26573-2_4. [DOI] [PubMed] [Google Scholar]
- 47.Buckley NE. The peripheral cannabinoid receptor knockout mice: an update. Br J Pharmacol. 2008;153:309–18. doi: 10.1038/sj.bjp.0707527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Beaulieu JM, Gainetdinov RR. The physiology, signaling, and pharmacology of dopamine receptors. Pharmacol Rev. 2011;63:182–217. doi: 10.1124/pr.110.002642. [DOI] [PubMed] [Google Scholar]
- 49.Neve KA, Seamans JK, Trantham-Davidson H. Dopamine receptor signaling. J Recept Signal Transduct Res. 2004;24:165–205. doi: 10.1081/rrs-200029981. [DOI] [PubMed] [Google Scholar]
- 50.Viggiano D, Ruocco LA, Sadile AG. Dopamine phenotype and behaviour in animal models: in relation to attention deficit hyperactivity disorder. Neurosci Biobehav Rev. 2003;27:623–37. doi: 10.1016/j.neubiorev.2003.08.006. [DOI] [PubMed] [Google Scholar]
- 51.Missale C, Nash SR, Robinson SW, Jaber M, Caron MG. Dopamine receptors: from structure to function. Physiol Rev. 1998;78:189–225. doi: 10.1152/physrev.1998.78.1.189. [DOI] [PubMed] [Google Scholar]
- 52.Drago J, Padungchaichot P, Accili D, Fuchs S. Dopamine receptors and dopamine transporter in brain function and addictive behaviors: insights from _targeted mouse mutants. Dev Neurosci. 1998;20:188–203. doi: 10.1159/000017313. [DOI] [PubMed] [Google Scholar]
- 53.Waddington JL, O’Tuathaigh C, O’Sullivan G, Tomiyama K, Koshikawa N, Croke DT. Phenotypic studies on dopamine receptor subtype and associated signal transduction mutants: insights and challenges from 10 years at the psychopharmacology-molecular biology interface. Psychopharmacology (Berl) 2005;181:611–38. doi: 10.1007/s00213-005-0058-8. [DOI] [PubMed] [Google Scholar]
- 54.Xu M, Hu XT, Cooper DC, Moratalla R, Graybiel AM, White FJ, et al. Elimination of cocaine-induced hyperactivity and dopamine-mediated neurophysiological effects in dopamine D1 receptor mutant mice. Cell. 1994;79:945–55. doi: 10.1016/0092-8674(94)90026-4. [DOI] [PubMed] [Google Scholar]
- 55.De Mei C, Ramos M, Iitaka C, Borrelli E. Getting specialized: presynaptic and post-synaptic dopamine D2 receptors. Curr Opin Pharmacol. 2009;9:53–8. doi: 10.1016/j.coph.2008.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Zhu J, Chen YJ, Zhao N, Cao GF, Dang YH, Han W, et al. Distinct roles of dopamine D3 receptors in modulating methamphetamine-induced behavioral sensitization and ultrastructural plasticity in the shell of the nucleus accumbens. J Neurosci Res. 2012;90:895–904. doi: 10.1002/jnr.22821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Harrison SJ, Nobrega JN. Differential susceptibility to ethanol and amphetamine sensitization in dopamine D3 receptor-deficient mice. Psychopharmacology (Berl) 2009;204:49–59. doi: 10.1007/s00213-008-1435-x. [DOI] [PubMed] [Google Scholar]
- 58.Li T, Hou Y, Cao W, Yan CX, Chen T, Li SB. Role of dopamine D3 receptors in basal nociception regulation and in morphine-induced tolerance and withdrawal. Brain Res. 2012;1433:80–4. doi: 10.1016/j.brainres.2011.11.045. [DOI] [PubMed] [Google Scholar]
- 59.Kaczor AA, Selent J. Oligomerization of G protein-coupled receptors: biochemical and biophysical methods. Curr Med Chem. 2011;18:4606–34. doi: 10.2174/092986711797379285. [DOI] [PubMed] [Google Scholar]
- 60.Tadagaki K, Jockers R, Kamal M. History and biological significance of GPCR heteromerization in the neuroendocrine system. Neuroendocrinology. 2012;95:223–31. doi: 10.1159/000330000. [DOI] [PubMed] [Google Scholar]
- 61.Gonzalez-Maeso J. GPCR oligomers in pharmacology and signaling. Mol Brain. 2011;4:20. doi: 10.1186/1756-6606-4-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Gomes I, Jordan BA, Gupta A, Rios C, Trapaidze N, Devi LA. G protein coupled receptor dimerization: implications in modulating receptor function. J Mol Med (Berl) 2001;79:226–42. doi: 10.1007/s001090100219. [DOI] [PubMed] [Google Scholar]
- 63.Rios CD, Jordan BA, Gomes I, Devi LA. G-protein-coupled receptor dimerization: modulation of receptor function. Pharmacol Ther. 2001;92:71–87. doi: 10.1016/s0163-7258(01)00160-7. [DOI] [PubMed] [Google Scholar]
- 64.Rozenfeld R, Devi LA. Exploring a role for heteromerization in GPCR signalling specificity. Biochem J. 2011;433:11–8. doi: 10.1042/BJ20100458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Rozenfeld R, Devi LA. Receptor heteromerization and drug discovery. Trends Pharmacol Sci. 2010;31:124–30. doi: 10.1016/j.tips.2009.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Rostene WH, Fischette CT, McEwen BS. Modulation by vasoactive intestinal peptide (VIP) of serotonin receptors in membranes from rat hippocampus. J Neurosci. 1983;3:2414–9. doi: 10.1523/JNEUROSCI.03-12-02414.1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Fuxe K, Ferre S, Zoli M, Agnati LF. Integrated events in central dopamine transmission as analyzed at multiple levels. Evidence for intramembrane adenosine A2A/dopamine D2 and adenosine A1/dopamine D1 receptor interactions in the basal ganglia. Brain Res Brain Res Rev. 1998;26:258–73. doi: 10.1016/s0165-0173(97)00049-0. [DOI] [PubMed] [Google Scholar]
- 68.Zoli M, Agnati LF, Hedlund PB, Li XM, Ferre S, Fuxe K. Receptor-receptor interactions as an integrative mechanism in nerve cells. Mol Neurobiol. 1993;7:293–334. doi: 10.1007/BF02769180. [DOI] [PubMed] [Google Scholar]
- 69.Fuxe K, Agnati LF, Benfenati F, Cimmino M, Algeri S, Hokfelt T, et al. Modulation by cholecystokinins of 3H-spiroperidol binding in rat striatum: evidence for increased affinity and reduction in the number of binding sites. Acta Physiol Scand. 1981;113:567–9. doi: 10.1111/j.1748-1716.1981.tb06942.x. [DOI] [PubMed] [Google Scholar]
- 70.Maggi A, U’Prichard DC, Enna SJ. beta-Adrenergic regulation of alpha 2-adrenergic receptors in the central nervous system. Science. 1980;207:645–7. doi: 10.1126/science.6101510. [DOI] [PubMed] [Google Scholar]
- 71.Maggio R, Vogel Z, Wess J. Coexpression studies with mutant muscarinic/adrenergic receptors provide evidence for intermolecular “cross-talk” between G-protein-linked receptors. Proc Natl Acad Sci USA. 1993;90:3103–7. doi: 10.1073/pnas.90.7.3103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Monnot C, Bihoreau C, Conchon S, Curnow KM, Corvol P, Clauser E. Polar residues in the transmembrane domains of the type 1 angiotensin II receptor are required for binding and coupling. Reconstitution of the binding site by co-expression of two deficient mutants. J Biol Chem. 1996;271:1507–13. doi: 10.1074/jbc.271.3.1507. [DOI] [PubMed] [Google Scholar]
- 73.Jones KA, Borowsky B, Tamm JA, Craig DA, Durkin MM, Dai M, et al. GABA(B) receptors function as a heteromeric assembly of the subunits GABA(B)R1 and GABA (B)R2. Nature. 1998;396:674–9. doi: 10.1038/25348. [DOI] [PubMed] [Google Scholar]
- 74.Kaupmann K, Malitschek B, Schuler V, Heid J, Froestl W, Beck P, et al. GABA(B)-receptor subtypes assemble into functional heteromeric complexes. Nature. 1998;396:683–7. doi: 10.1038/25360. [DOI] [PubMed] [Google Scholar]
- 75.Kuner R, Kohr G, Grunewald S, Eisenhardt G, Bach A, Kornau HC. Role of heteromer formation in GABAB receptor function. Science. 1999;283:74–7. doi: 10.1126/science.283.5398.74. [DOI] [PubMed] [Google Scholar]
- 76.White JH, Wise A, Main MJ, Green A, Fraser NJ, Disney GH, et al. Heterodimerization is required for the formation of a functional GABA(B) receptor. Nature. 1998;396:679–82. doi: 10.1038/25354. [DOI] [PubMed] [Google Scholar]
- 77.Cui M, Jiang P, Maillet E, Max M, Margolskee RF, Osman R. The heterodimeric sweet taste receptor has multiple potential ligand binding sites. Curr Pharm Des. 2006;12:4591–600. doi: 10.2174/138161206779010350. [DOI] [PubMed] [Google Scholar]
- 78.Nelson G, Hoon MA, Chandrashekar J, Zhang Y, Ryba NJ, Zuker CS. Mammalian sweet taste receptors. Cell. 2001;106:381–90. doi: 10.1016/s0092-8674(01)00451-2. [DOI] [PubMed] [Google Scholar]
- 79.Nelson G, Chandrashekar J, Hoon MA, Feng L, Zhao G, Ryba NJ, et al. An aminoacid taste receptor. Nature. 2002;416:199–202. doi: 10.1038/nature726. [DOI] [PubMed] [Google Scholar]
- 80.Gomes I, Filipovska J, Jordan BA, Devi LA. Oligomerization of opioid receptors. Methods. 2002;27:358–65. doi: 10.1016/s1046-2023(02)00094-4. [DOI] [PubMed] [Google Scholar]
- 81.Gomes I, Filipovska J, Devi LA. Opioid receptor oligomerization. Detection and functional characterization of interacting receptors. Methods Mol Med. 2003;84:157–83. doi: 10.1385/1-59259-379-8:157. [DOI] [PubMed] [Google Scholar]
- 82.Jordan BA, Devi LA. G-protein-coupled receptor heterodimerization modulates receptor function. Nature. 1999;399:697–700. doi: 10.1038/21441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Salahpour A, Espinoza S, Masri B, Lam V, Barak LS, Gainetdinov RR. BRET biosensors to study GPCR biology, pharmacology, and signal transduction. Front Endocrinol (Lausanne) 2012;3:105. doi: 10.3389/fendo.2012.00105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Overland AC, Kitto KF, Chabot-Dore AJ, Rothwell PE, Fairbanks CA, Stone LS, et al. Protein kinase C mediates the synergistic interaction between agonists acting at alpha(2)-adrenergic and delta-opioid receptors in spinal cord. J Neurosci. 2009;29:13264–73. doi: 10.1523/JNEUROSCI.1907-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Rios C, Gomes I, Devi LA. Interactions between delta opioid receptors and alpha-adrenoceptors. Clin Exp Pharmacol Physiol. 2004;31:833–6. doi: 10.1111/j.1440-1681.2004.04076.x. [DOI] [PubMed] [Google Scholar]
- 86.Ramsay D, Kellett E, McVey M, Rees S, Milligan G. Homo- and hetero-oligomeric interactions between G-protein-coupled receptors in living cells monitored by two variants of bioluminescence resonance energy transfer (BRET): hetero-oligomers between receptor subtypes form more efficiently than between less closely related sequences. Biochem J. 2002;365:429–40. doi: 10.1042/BJ20020251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Jordan BA, Trapaidze N, Gomes I, Nivarthi R, Devi LA. Oligomerization of opioid receptors with beta 2-adrenergic receptors: a role in trafficking and mitogen-activated protein kinase activation. Proc Natl Acad Sci USA. 2001;98:343–8. doi: 10.1073/pnas.011384898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Rozenfeld R, Bushlin I, Gomes I, Tzavaras N, Gupta A, Neves S, et al. Receptor heteromerization expands the repertoire of cannabinoid signaling in rodent neurons. PLoS One. 2012;7:e29239. doi: 10.1371/journal.pone.0029239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Rios C, Gomes I, Devi LA. Mu opioid and CB1 cannabinoid receptor interactions: reciprocal inhibition of receptor signaling and neuritogenesis. Br J Pharmacol. 2006;148:387–95. doi: 10.1038/sj.bjp.0706757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Pello OM, Martinez-Munoz L, Parrillas V, Serrano A, Rodriguez-Frade JM, Toro MJ, et al. Ligand stabilization of CXCR4/delta-opioid receptor heterodimers reveals a mechanism for immune response regulation. Eur J Immunol. 2008;38:537–49. doi: 10.1002/eji.200737630. [DOI] [PubMed] [Google Scholar]
- 91.Ambrose-Lanci LM, Peiris NB, Unterwald EM, Van Bockstaele EJ. Cocaine withdrawal-induced trafficking of delta-opioid receptors in rat nucleus accumbens. Brain Res. 2008;1210:92–102. doi: 10.1016/j.brainres.2008.02.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Ambrose LM, Gallagher SM, Unterwald EM, Van Bockstaele EJ. Dopamine-D1 and delta-opioid receptors co-exist in rat striatal neurons. Neurosci Lett. 2006;399:191–6. doi: 10.1016/j.neulet.2006.02.027. [DOI] [PubMed] [Google Scholar]
- 93.Gomes I, Jordan BA, Gupta A, Trapaidze N, Nagy V, Devi LA. Heterodimerization of mu and delta opioid receptors: a role in opiate synergy. J Neurosci. 2000;20:RC110. doi: 10.1523/JNEUROSCI.20-22-j0007.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Gomes I, Gupta A, Filipovska J, Szeto HH, Pintar JE, Devi LA. A role for heterodimerization of mu and delta opiate receptors in enhancing morphine analgesia. Proc Natl Acad Sci USA. 2004;101:5135–9. doi: 10.1073/pnas.0307601101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.George SR, Fan T, Xie Z, Tse R, Tam V, Varghese G, et al. Oligomerization of mu-and delta-opioid receptors. Generation of novel functional properties. J Biol Chem. 2000;275:26128–35. doi: 10.1074/jbc.M000345200. [DOI] [PubMed] [Google Scholar]
- 96.Gomes I, Ijzerman AP, Ye K, Maillet EL, Devi LA. G protein-coupled receptor heteromerization: a role in allosteric modulation of ligand binding. Mol Pharmacol. 2011;79:1044–52. doi: 10.1124/mol.110.070847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Kabli N, Martin N, Fan T, Nguyen T, Hasbi A, Balboni G, et al. Agonists at the delta-opioid receptor modify the binding of micro-receptor agonists to the micro-delta receptor hetero-oligomer. Br J Pharmacol. 2010;161:1122–36. doi: 10.1111/j.1476-5381.2010.00944.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Fan T, Varghese G, Nguyen T, Tse R, O’Dowd BF, George SR. A role for the distal carboxyl tails in generating the novel pharmacology and G protein activation profile of mu and delta opioid receptor hetero-oligomers. J Biol Chem. 2005;280:38478–88. doi: 10.1074/jbc.M505644200. [DOI] [PubMed] [Google Scholar]
- 99.Hasbi A, Nguyen T, Fan T, Cheng R, Rashid A, Alijaniaram M, et al. Trafficking of preassembled opioid mu-delta heterooligomer-Gz signaling complexes to the plasma membrane: coregulation by agonists. Biochemistry. 2007;46:12997–3009. doi: 10.1021/bi701436w. [DOI] [PubMed] [Google Scholar]
- 100.Rozenfeld R, Devi LA. Receptor heterodimerization leads to a switch in signaling: beta-arrestin2-mediated ERK activation by mu-delta opioid receptor heterodimers. FASEB J. 2007;21:2455–65. doi: 10.1096/fj.06-7793com. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Charles AC, Mostovskaya N, Asas K, Evans CJ, Dankovich ML, Hales TG. Coexpression of delta-opioid receptors with micro receptors in GH3 cells changes the functional response to micro agonists from inhibitory to excitatory. Mol Pharmacol. 2003;63:89–95. doi: 10.1124/mol.63.1.89. [DOI] [PubMed] [Google Scholar]
- 102.Law PY, Erickson-Herbrandson LJ, Zha QQ, Solberg J, Chu J, Sarre A, et al. Heterodimerization of mu- and delta-opioid receptors occurs at the cell surface only and requires receptor-G protein interactions. J Biol Chem. 2005;280:11152–64. doi: 10.1074/jbc.M500171200. [DOI] [PubMed] [Google Scholar]
- 103.Milan-Lobo L, Whistler JL. Heteromerization of the mu- and delta-opioid receptors produces ligand-biased antagonism and alters mu-receptor trafficking. J Pharmacol Exp Ther. 2011;337:868–75. doi: 10.1124/jpet.111.179093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Breit A, Gagnidze K, Devi LA, Lagace M, Bouvier M. Simultaneous activation of the delta opioid receptor (deltaOR)/sensory neuron-specific receptor-4 (SNSR-4) heterooligomer by the mixed bivalent agonist bovine adrenal medulla peptide 22 activates SNSR-4 but inhibits deltaOR signaling. Mol Pharmacol. 2006;70:686–96. doi: 10.1124/mol.106.022897. [DOI] [PubMed] [Google Scholar]
- 105.Berrendero F, Maldonado R. Involvement of the opioid system in the anxiolytic-like effects induced by Delta(9)-tetrahydrocannabinol. Psychopharmacology (Berl) 2002;163:111–7. doi: 10.1007/s00213-002-1144-9. [DOI] [PubMed] [Google Scholar]
- 106.Li Y, Chen J, Bai B, Du H, Liu Y, Liu H. Heterodimerization of human apelin and kappa opioid receptors: roles in signal transduction. Cell Signal. 2012;24:991–1001. doi: 10.1016/j.cellsig.2011.12.012. [DOI] [PubMed] [Google Scholar]
- 107.Jordan BA, Gomes I, Rios C, Filipovska J, Devi LA. Functional interactions between mu opioid and alpha 2A-adrenergic receptors. Mol Pharmacol. 2003;64:1317–24. doi: 10.1124/mol.64.6.1317. [DOI] [PubMed] [Google Scholar]
- 108.Vilardaga JP, Nikolaev VO, Lorenz K, Ferrandon S, Zhuang Z, Lohse MJ. Conformational cross-talk between alpha2A-adrenergic and mu-opioid receptors controls cell signaling. Nat Chem Biol. 2008;4:126–31. doi: 10.1038/nchembio.64. [DOI] [PubMed] [Google Scholar]
- 109.Zhang YQ, Limbird LE. Hetero-oligomers of alpha2A-adrenergic and mu-opioid receptors do not lead to transactivation of G-proteins or altered endocytosis profiles. Biochem Soc Trans. 2004;32:856–60. doi: 10.1042/BST0320856. [DOI] [PubMed] [Google Scholar]
- 110.Hojo M, Sudo Y, Ando Y, Minami K, Takada M, Matsubara T, et al. mu-Opioid receptor forms a functional heterodimer with cannabinoid CB1 receptor: electrophysiological and FRET assay analysis. J Pharmacol Sci. 2008;108:308–19. doi: 10.1254/jphs.08244fp. [DOI] [PubMed] [Google Scholar]
- 111.Chen CG, Li J, Bot G, Szabo I, Rogers TJ, Liu-Chen LY. Heterodimerization and cross-desensitization between the mu-opioid receptor and the chemokine CCR5 receptor. Eur J Pharmacol. 2004;483:175–86. doi: 10.1016/j.ejphar.2003.10.033. [DOI] [PubMed] [Google Scholar]
- 112.Suzuki S, Chuang LF, Yau P, Doi RH, Chuang RY. Interactions of opioid and chemokine receptors: oligomerization of mu, kappa, and delta with CCR5 on immune cells. Exp Cell Res. 2002;280:192–200. doi: 10.1006/excr.2002.5638. [DOI] [PubMed] [Google Scholar]
- 113.Liu XY, Liu ZC, Sun YG, Ross M, Kim S, Tsai FF, et al. Unidirectional cross-activation of GRPR by MOR1D uncouples itch and analgesia induced by opioids. Cell. 2011;147:447–58. doi: 10.1016/j.cell.2011.08.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Wang D, Sun X, Bohn LM, Sadee W. Opioid receptor homo- and heterodimerization in living cells by quantitative bioluminescence resonance energy transfer. Mol Pharmacol. 2005;67:2173–84. doi: 10.1124/mol.104.010272. [DOI] [PubMed] [Google Scholar]
- 115.Chakrabarti S, Liu NJ, Gintzler AR. Formation of mu-/kappa-opioid receptor heterodimer is sex-dependent and mediates female-specific opioid analgesia. Proc Natl Acad Sci USA. 2010;107:20115–9. doi: 10.1073/pnas.1009923107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Pfeiffer M, Kirscht S, Stumm R, Koch T, Wu DF, Laugsch M, et al. Heterodimerization of substance P and mu-opioid receptors regulates receptor trafficking and resensitization. J Biol Chem. 2003;278:51630–7. doi: 10.1074/jbc.M307095200. [DOI] [PubMed] [Google Scholar]
- 117.Pan YX, Bolan E, Pasternak GW. Dimerization of morphine and orphanin FQ/nociceptin receptors: generation of a novel opioid receptor subtype. Biochem Biophys Res Commun. 2002;297:659–63. doi: 10.1016/s0006-291x(02)02258-1. [DOI] [PubMed] [Google Scholar]
- 118.Wang HL, Hsu CY, Huang PC, Kuo YL, Li AH, Yeh TH, et al. Heterodimerization of opioid receptor-like 1 and mu-opioid receptors impairs the potency of micro receptor agonist. J Neurochem. 2005;92:1285–94. doi: 10.1111/j.1471-4159.2004.02921.x. [DOI] [PubMed] [Google Scholar]
- 119.Pfeiffer M, Koch T, Schroder H, Laugsch M, Hollt V, Schulz S. Heterodimerization of somatostatin and opioid receptors cross-modulates phosphorylation, internalization, and desensitization. J Biol Chem. 2002;277:19762–72. doi: 10.1074/jbc.M110373200. [DOI] [PubMed] [Google Scholar]
- 120.Gell DA, Grant RP, Mackay JP. The detection and quantitation of protein oligomerization. Adv Exp Med Biol. 2012;747:19–41. doi: 10.1007/978-1-4614-3229-6_2. [DOI] [PubMed] [Google Scholar]
- 121.Ciruela F, Vilardaga JP, Fernandez-Duenas V. Lighting up multiprotein complexes: lessons from GPCR oligomerization. Trends Biotechnol. 2010;28:407–15. doi: 10.1016/j.tibtech.2010.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Day RN, Davidson MW. Fluorescent proteins for FRET microscopy: monitoring protein interactions in living cells. Bioessays. 2012;34:341–50. doi: 10.1002/bies.201100098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Padilla-Parra S, Tramier M. FRET microscopy in the living cell: different approaches, strengths and weaknesses. Bioessays. 2012;34:369–76. doi: 10.1002/bies.201100086. [DOI] [PubMed] [Google Scholar]
- 124.Lohse MJ, Nuber S, Hoffmann C. Fluorescence/bioluminescence resonance energy transfer techniques to study G-protein-coupled receptor activation and signaling. Pharmacol Rev. 2012;64:299–336. doi: 10.1124/pr.110.004309. [DOI] [PubMed] [Google Scholar]
- 125.Urizar E, Yano H, Kolster R, Gales C, Lambert N, Javitch JA. CODA-RET reveals functional selectivity as a result of GPCR heteromerization. Nat Chem Biol. 2011;7:624–30. doi: 10.1038/nchembio.623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Dragulescu-Andrasi A, Chan CT, De A, Massoud TF, Gambhir SS. Bioluminescence resonance energy transfer (BRET) imaging of protein-protein interactions within deep tissues of living subjects. Proc Natl Acad Sci USA. 2011;108:12060–5. doi: 10.1073/pnas.1100923108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Cabello N, Gandia J, Bertarelli DC, Watanabe M, Lluis C, Franco R, et al. Metabotropic glutamate type 5, dopamine D2 and adenosine A2a receptors form higher-order oligomers in living cells. J Neurochem. 2009;109:1497–507. doi: 10.1111/j.1471-4159.2009.06078.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Gandia J, Galino J, Amaral OB, Soriano A, Lluis C, Franco R, et al. Detection of higher-order G protein-coupled receptor oligomers by a combined BRET-BiFC technique. FEBS Lett. 2008;582:2979–84. doi: 10.1016/j.febslet.2008.07.045. [DOI] [PubMed] [Google Scholar]
- 129.Navarro G, Carriba P, Gandia J, Ciruela F, Casado V, Cortes A, et al. Detection of heteromers formed by cannabinoid CB1, dopamine D2, and adenosine A2A G-protein-coupled receptors by combining bimolecular fluorescence complementation and bioluminescence energy transfer. Scientific World J. 2008;8:1088–97. doi: 10.1100/tsw.2008.136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Vidi PA, Chen JJ, Irudayaraj JMK, Watts VJ. Adenosine A(2A) receptors assemble into higher-order oligomers at the plasma membrane. FEBS Lett. 2008;582:3985–90. doi: 10.1016/j.febslet.2008.09.062. [DOI] [PubMed] [Google Scholar]
- 131.Binnig G, Quate CF, Gerber C. Atomic force microscope. Phys Rev Lett. 1986;56:930–3. doi: 10.1103/PhysRevLett.56.930. [DOI] [PubMed] [Google Scholar]
- 132.Fotiadis D, Liang Y, Filipek S, Saperstein DA, Engel A, Palczewski K. The G protein-coupled receptor rhodopsin in the native membrane. FEBS Lett. 2004;564:281–8. doi: 10.1016/S0014-5793(04)00194-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Kunishima N, Shimada Y, Tsuji Y, Sato T, Yamamoto M, Kumasaka T, et al. Structural basis of glutamate recognition by a dimeric metabotropic glutamate receptor. Nature. 2000;407:971–7. doi: 10.1038/35039564. [DOI] [PubMed] [Google Scholar]
- 134.Fotiadis D, Jastrzebska B, Philippsen A, Muller DJ, Palczewski K, Engel A. Structure of the rhodopsin dimer: a working model for G-protein-coupled receptors. Curr Opin Struct Biol. 2006;16:252–9. doi: 10.1016/j.sbi.2006.03.013. [DOI] [PubMed] [Google Scholar]
- 135.Cherezov V, Rosenbaum DM, Hanson MA, Rasmussen SG, Thian FS, Kobilka TS, et al. High-resolution crystal structure of an engineered human beta2-adrenergic G protein-coupled receptor. Science. 2007;318:1258–65. doi: 10.1126/science.1150577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Hanson MA, Stevens RC. Discovery of new GPCR biology: one receptor structure at a time. Structure. 2009;17:8–14. doi: 10.1016/j.str.2008.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Wu BL, Chien EYT, Mol CD, Fenalti G, Liu W, Katritch V, et al. Structures of the CXCR4 chemokine GPCR with small-molecule and cyclic peptide antagonists. Science. 2010;330:1066–71. doi: 10.1126/science.1194396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Chien EYT, Liu W, Zhao QA, Katritch V, Han GW, Hanson MA, et al. Structure of the human dopamine D3 receptor in complex with a D2/D3 selective antagonist. Science. 2010;330:1091–5. doi: 10.1126/science.1197410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Manglik A, Kruse AC, Kobilka TS, Thian FS, Mathiesen JM, Sunahara RK, et al. Crystal structure of the micro-opioid receptor bound to a morphinan antagonist. Nature. 2012;485:321–6. doi: 10.1038/nature10954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Wu H, Wacker D, Mileni M, Katritch V, Han GW, Vardy E, et al. Structure of the human kappa-opioid receptor in complex with JDTic. Nature. 2012;485:327–32. doi: 10.1038/nature10939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Granier S, Manglik A, Kruse AC, Kobilka TS, Thian FS, Weis WI, et al. Structure of the delta-opioid receptor bound to naltrindole. Nature. 2012;485:400–4. doi: 10.1038/nature11111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Thompson AA, Liu W, Chun E, Katritch V, Wu H, Vardy E, et al. Structure of the nociceptin/orphanin FQ receptor in complex with a peptide mimetic. Nature. 2012;485:395–9. doi: 10.1038/nature11085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Albizu L, Cottet M, Kralikova M, Stoev S, Seyer R, Brabet I, et al. Time-resolved FRET between GPCR ligands reveals oligomers in native tissues. Nat Chem Biol. 2010;6:587–94. doi: 10.1038/nchembio.396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Rocheville M, Lange DC, Kumar U, Patel SC, Patel RC, Patel YC. Receptors for dopamine and somatostatin: formation of hetero-oligomers with enhanced functional activity. Science. 2000;288:154–7. doi: 10.1126/science.288.5463.154. [DOI] [PubMed] [Google Scholar]
- 145.Rocheville M, Lange DC, Kumar U, Sasi R, Patel RC, Patel YC. Subtypes of the somatostatin receptor assemble as functional homo- and heterodimers. J Biol Chem. 2000;275:7862–9. doi: 10.1074/jbc.275.11.7862. [DOI] [PubMed] [Google Scholar]
- 146.McVey M, Ramsay D, Kellett E, Rees S, Wilson S, Pope AJ, et al. Monitoring receptor oligomerization using time-resolved fluorescence resonance energy transfer and bioluminescence resonance energy transfer—the human delta-opioid receptor displays constitutive oligomerization at the cell surface, which is not regulated by receptor occupancy. J Biol Chem. 2001;276:14092–9. doi: 10.1074/jbc.M008902200. [DOI] [PubMed] [Google Scholar]
- 147.Comps-Agrar L, Kniazeff J, Norskov-Lauritsen L, Maurel D, Gassmann M, Gregor N, et al. The oligomeric state sets GABA(B) receptor signalling efficacy. EMBO J. 2011;30:2336–49. doi: 10.1038/emboj.2011.143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Callen L, Moreno E, Barroso-Chinea P, Moreno-Delgado D, Cortes A, Mallol J, et al. Cannabinoid receptors CB1 and CB2 form functional heteromers in brain. J Biol Chem. 2012;287:20851–65. doi: 10.1074/jbc.M111.335273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Trifilieff P, Rives ML, Urizar E, Piskorowski RA, Vishwasrao HD, Castrillon J, et al. Detection of antigen interactions ex vivo by proximity ligation assay: endogenous dopamine D2-adenosine A2A receptor complexes in the striatum. Biotechniques. 2011;51:111–8. doi: 10.2144/000113719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Jiang Q, Takemori AE, Sultana M, Portoghese PS, Bowen WD, Mosberg HI, et al. Differential antagonism of opioid delta antinociception by [D-Ala2, Leu5, Cys6] enkephalin and naltrindole 5′-isothiocyanate: evidence for delta receptor subtypes. J Pharmacol Exp Ther. 1991;257:1069–75. [PubMed] [Google Scholar]
- 151.Zukin RS, Eghbali M, Olive D, Unterwald EM, Tempel A. Characterization and visualization of rat and guinea pig brain kappa opioid receptors: evidence for kappa 1 and kappa 2 opioid receptors. Proc Natl Acad Sci USA. 1988;85:4061–5. doi: 10.1073/pnas.85.11.4061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Bhushan RG, Sharma SK, Xie Z, Daniels DJ, Portoghese PS. A bivalent ligand (KDN-21) reveals spinal delta and kappa opioid receptors are organized as heterodimers that give rise to delta(1) and kappa(2) phenotypes. Selective _targeting of delta-kappa heterodimers. J Med Chem. 2004;47:2969–72. doi: 10.1021/jm0342358. [DOI] [PubMed] [Google Scholar]
- 153.Xie Z, Bhushan RG, Daniels DJ, Portoghese PS. Interaction of bivalent ligand KDN21 with heterodimeric delta-kappa opioid receptors in human embryonic kidney 293 cells. Mol Pharmacol. 2005;68:1079–86. doi: 10.1124/mol.105.012070. [DOI] [PubMed] [Google Scholar]
- 154.Waldhoer M, Fong J, Jones RM, Lunzer MM, Sharma SK, Kostenis E, et al. A heterodimer-selective agonist shows in vivo relevance of G protein-coupled receptor dimers. Proc Natl Acad Sci USA. 2005;102:9050–5. doi: 10.1073/pnas.0501112102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Rives ML, Rossillo M, Liu-Chen LY, Javitch JA. 6′-Guanidinonaltrindole (6′-GNTI) is a G protein-biased kappa-opioid receptor agonist that inhibits arrestin recruitment. J Biol Chem. 2012;287:27050–4. doi: 10.1074/jbc.C112.387332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Costantino CM, Gomes I, Stockton SD, Lim MP, Devi LA. Opioid receptor heteromers in analgesia. Expert Rev Mol Med. 2012:14. doi: 10.1017/erm.2012.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Rothman RB, Bowen WD, Herkenham M, Jacobson AE, Rice KC, Pert CB. A quantitative study of [3H]D-Ala2-D-Leu5-enkephalin binding to rat brain membranes. Evidence that oxymorphone is a noncompetitive inhibitor of the lower affinity delta-binding site. Mol Pharmacol. 1985;27:399–409. [PubMed] [Google Scholar]
- 158.Rothman RB, Long JB, Bykov V, Jacobson AE, Rice KC, Holaday JW. Beta-FNA binds irreversibly to the opiate receptor complex: in vivo and in vitro evidence. J Pharmacol Exp Ther. 1988;247:405–16. [PubMed] [Google Scholar]
- 159.Porreca F, Takemori AE, Sultana M, Portoghese PS, Bowen WD, Mosberg HI. Modulation of mu-mediated antinociception in the mouse involves opioid delta-2 receptors. J Pharmacol Exp Ther. 1992;263:147–52. [PubMed] [Google Scholar]
- 160.Abul-Husn NS, Sutak M, Milne B, Jhamandas K. Augmentation of spinal morphine analgesia and inhibition of tolerance by low doses of mu- and delta-opioid receptor antagonists. Br J Pharmacol. 2007;151:877–87. doi: 10.1038/sj.bjp.0707277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Abdelhamid EE, Takemori AE. Characteristics of mu and delta opioid binding sites in striatal slices of morphine-tolerant and -dependent mice. Eur J Pharmacol. 1991;198:157–63. doi: 10.1016/0014-2999(91)90615-w. [DOI] [PubMed] [Google Scholar]
- 162.Zhu Y, King MA, Schuller AG, Nitsche JF, Reidl M, Elde RP, et al. Retention of supraspinal delta-like analgesia and loss of morphine tolerance in delta opioid receptor knockout mice. Neuron. 1999;24:243–52. doi: 10.1016/s0896-6273(00)80836-3. [DOI] [PubMed] [Google Scholar]
- 163.Shapira M, Gafni M, Sarne Y. Independence of, and interactions between, cannabinoid and opioid signal transduction pathways in N18TG2 cells. Brain Res. 1998;806:26–35. doi: 10.1016/s0006-8993(98)00697-0. [DOI] [PubMed] [Google Scholar]
- 164.Berrendero F, Mendizabal V, Murtra P, Kieffer BL, Maldonado R. Cannabinoid receptor and WIN 55 212-2-stimulated [35S]-GTPgammaS binding in the brain of mu-, delta- and kappa-opioid receptor knockout mice. Eur J Neurosci. 2003;18:2197–202. doi: 10.1046/j.1460-9568.2003.02951.x. [DOI] [PubMed] [Google Scholar]
- 165.Uriguen L, Berrendero F, Ledent C, Maldonado R, Manzanares J. Kappa- and delta-opioid receptor functional activities are increased in the caudate putamen of cannabinoid CB1 receptor knockout mice. Eur J Neurosci. 2005;22:2106–10. doi: 10.1111/j.1460-9568.2005.04372.x. [DOI] [PubMed] [Google Scholar]
- 166.Grazzini E, Puma C, Roy MO, Yu XH, O’Donnell D, Schmidt R, et al. Sensory neuron-specific receptor activation elicits central and peripheral nociceptive effects in rats. Proc Natl Acad Sci USA. 2004;101:7175–80. doi: 10.1073/pnas.0307185101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Lembo PM, Grazzini E, Groblewski T, O’Donnell D, Roy MO, Zhang J, et al. Proenkephalin A gene products activate a new family of sensory neuron-specific GPCRs. Nat Neurosci. 2002;5:201–9. doi: 10.1038/nn815. [DOI] [PubMed] [Google Scholar]
- 168.Pepe S, Xiao RP, Hohl C, Altschuld R, Lakatta EG. ‘Cross talk’ between opioid peptide and adrenergic receptor signaling in isolated rat heart. Circulation. 1997;95:2122–9. doi: 10.1161/01.cir.95.8.2122. [DOI] [PubMed] [Google Scholar]
- 169.Xiao RP, Pepe S, Spurgeon HA, Capogrossi MC, Lakatta EG. Opioid peptide receptor stimulation reverses beta-adrenergic effects in rat heart cells. Am J Physiol Heart Circ Physiol. 1997;272:H797–H805. doi: 10.1152/ajpheart.1997.272.2.H797. [DOI] [PubMed] [Google Scholar]
- 170.Fairbanks CA, Posthumus IJ, Kitto KF, Stone LS, Wilcox GL. Moxonidine, a selective imidazoline/alpha(2) adrenergic receptor agonist, synergizes with morphine and deltorphin II to inhibit substance P-induced behavior in mice. Pain. 2000;84:13–20. doi: 10.1016/S0304-3959(99)00171-2. [DOI] [PubMed] [Google Scholar]
- 171.Fairbanks CA, Stone LS, Kitto KF, Nguyen HO, Posthumus IJ, Wilcox GL. alpha(2C)-Adrenergic receptors mediate spinal analgesia and adrenergic-opioid synergy. J Pharmacol Exp Ther. 2002;300:282–90. doi: 10.1124/jpet.300.1.282. [DOI] [PubMed] [Google Scholar]
- 172.Guo XH, Fairbanks CA, Stone LS, Loh HH. DPDPE-UK14,304 synergy is retained in mu opioid receptor knockout mice. Pain. 2003;104:209–17. doi: 10.1016/s0304-3959(03)00007-1. [DOI] [PubMed] [Google Scholar]
- 173.Stone LS, MacMillan LB, Kitto KF, Limbird LE, Wilcox GL. The alpha2a adrenergic receptor subtype mediates spinal analgesia evoked by alpha2 agonists and is necessary for spinal adrenergic-opioid synergy. J Neurosci. 1997;17:7157–65. doi: 10.1523/JNEUROSCI.17-18-07157.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Riedl MS, Schnell SA, Overland AC, Chabot-Dore AJ, Taylor AM, Ribeiro-Da-Silva A, et al. Coexpression of alpha(2A)-adrenergic and delta-opioid receptors in substance P-containing terminals in rat dorsal horn (vol. 513, p. 385, 2009) J Comp Neurol. 2009;514:674 674. doi: 10.1002/cne.21982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Daunais JB, McGinty JF. Acute and chronic cocaine administration differentially alters striatal opioid and nuclear transcription factor mRNAs. Synapse. 1994;18:35–45. doi: 10.1002/syn.890180106. [DOI] [PubMed] [Google Scholar]
- 176.George SR, Kertesz M. Met-enkephalin concentrations in striatum respond reciprocally to alterations in dopamine neurotransmission. Peptides. 1987;8:487–92. doi: 10.1016/0196-9781(87)90014-3. [DOI] [PubMed] [Google Scholar]
- 177.Hurd YL, Brown EE, Finlay JM, Fibiger HC, Gerfen CR. Cocaine self-administration differentially alters mRNA expression of striatal peptides. Brain Res Mol Brain Res. 1992;13:165–70. doi: 10.1016/0169-328x(92)90058-j. [DOI] [PubMed] [Google Scholar]
- 178.Unterwald EM, Cuntapay M. Dopamine-opioid interactions in the rat striatum: a modulatory role for dopamine D1 receptors in delta opioid receptor-mediated signal transduction. Neuropharmacology. 2000;39:372–81. doi: 10.1016/s0028-3908(99)00154-9. [DOI] [PubMed] [Google Scholar]
- 179.Ito S, Mori T, Sawaguchi T. Differential effects of micro-opioid, delta-opioid and kappa-opioid receptor agonists on dopamine receptor agonist-induced climbing behavior in mice. Behav Pharmacol. 2006;17:691–701. doi: 10.1097/FBP.0b013e32801155a1. [DOI] [PubMed] [Google Scholar]
- 180.Drasner K, Fields HL. Synergy between the antinociceptive effects of intrathecal clonidine and systemic morphine in the rat. Pain. 1988;32:309–12. doi: 10.1016/0304-3959(88)90042-5. [DOI] [PubMed] [Google Scholar]
- 181.Ossipov MH, Lopez Y, Bian D, Nichols ML, Porreca F. Synergistic antinociceptive interactions of morphine and clonidine in rats with nerve-ligation injury. Anesthesiology. 1997;86:196–204. doi: 10.1097/00000542-199701000-00024. [DOI] [PubMed] [Google Scholar]
- 182.Ledent C, Valverde O, Cossu G, Petitet F, Aubert JF, Beslot F, et al. Unresponsiveness to cannabinoids and reduced addictive effects of opiates in CB1 receptor knockout mice. Science. 1999;283:401–4. doi: 10.1126/science.283.5400.401. [DOI] [PubMed] [Google Scholar]
- 183.Chuang TK, Killam KF, Jr, Chuang LF, Kung HF, Sheng WS, Chao CC, et al. Mu opioid receptor gene expression in immune cells. Biochem Biophys Res Commun. 1995;216:922–30. doi: 10.1006/bbrc.1995.2709. [DOI] [PubMed] [Google Scholar]
- 184.Miyagi T, Chuang LF, Doi RH, Carlos MP, Torres JV, Chuang RY. Morphine induces gene expression of CCR5 in human CEM x174 lymphocytes. J Biol Chem. 2000;275:31305–10. doi: 10.1074/jbc.M001269200. [DOI] [PubMed] [Google Scholar]
- 185.Ballantyne JC, Loach AB, Carr DB. Itching after epidural and spinal opiates. Pain. 1988;33:149–60. doi: 10.1016/0304-3959(88)90085-1. [DOI] [PubMed] [Google Scholar]
- 186.Chaney MA. Side effects of intrathecal and epidural opioids. Can J Anaesth. 1995;42:891–903. doi: 10.1007/BF03011037. [DOI] [PubMed] [Google Scholar]
- 187.Hales P. Pruritus after epidural morphine. Lancet. 1980;2:204. doi: 10.1016/s0140-6736(80)90090-2. [DOI] [PubMed] [Google Scholar]
- 188.Sun YG, Chen ZF. A gastrin-releasing peptide receptor mediates the itch sensation in the spinal cord. Nature. 2007;448:700–3. doi: 10.1038/nature06029. [DOI] [PubMed] [Google Scholar]
- 189.Sun YG, Zhao ZQ, Meng XL, Yin J, Liu XY, Chen ZF. Cellular basis of itch sensation. Science. 2009;325:1531–4. doi: 10.1126/science.1174868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Rizzi A, Bigoni R, Marzola G, Guerrini R, Salvadori S, Regoli D, et al. The nociceptin/orphanin FQ receptor antagonist, [Nphe1]NC(1–13)NH2, potentiates morphine analgesia. Neuroreport. 2000;11:2369–72. doi: 10.1097/00001756-200008030-00007. [DOI] [PubMed] [Google Scholar]
- 191.King M, Chang A, Pasternak GW. Functional blockade of opioid analgesia by orphanin FQ/nociceptin. Biochem Pharmacol. 1998;55:1537–40. doi: 10.1016/s0006-2952(98)00023-9. [DOI] [PubMed] [Google Scholar]
- 192.Aicher SA, Punnoose A, Goldberg A. mu-Opioid receptors often colocalize with the substance P receptor (NK1) in the trigeminal dorsal horn. J Neurosci. 2000;20:4345–54. doi: 10.1523/JNEUROSCI.20-11-04345.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Aicher SA, Sharma S, Cheng PY, Liu-Chen LY, Pickel VM. Dual ultrastructural localization of mu-opiate receptors and substance P in the dorsal horn. Synapse. 2000;36:12–20. doi: 10.1002/(SICI)1098-2396(200004)36:1<12::AID-SYN2>3.0.CO;2-E. [DOI] [PubMed] [Google Scholar]
- 194.Wan Q, Douglas SD, Wang X, Kolson DL, O’Donnell LA, Ho WZ. Morphine upregulates functional expression of neurokinin-1 receptor in neurons. J Neurosci Res. 2006;84:1588–96. doi: 10.1002/jnr.21053. [DOI] [PubMed] [Google Scholar]
- 195.Yu YJ, Arttamangkul S, Evans CJ, Williams JT, von Zastrow M. Neurokinin 1 receptors regulate morphine-induced endocytosis and desensitization of mu-opioid receptors in CNS neurons. J Neurosci. 2009;29:222–33. doi: 10.1523/JNEUROSCI.4315-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Murtra P, Sheasby AM, Hunt SP, De Felipe C. Rewarding effects of opiates are absent in mice lacking the receptor for substance P. Nature. 2000;405:180–3. doi: 10.1038/35012069. [DOI] [PubMed] [Google Scholar]
- 197.Tai KK, Jin WQ, Chan TK, Wong TM. Characterization of [3H]U69593 binding sites in the rat heart by receptor binding assays. J Mol Cell Cardiol. 1991;23:1297–302. doi: 10.1016/0022-2828(91)90086-2. [DOI] [PubMed] [Google Scholar]
- 198.Ventura C, Bastagli L, Bernardi P, Caldarera CM, Guarnieri C. Opioid receptors in rat cardiac sarcolemma: effect of phenylephrine and isoproterenol. Biochim Biophys Acta. 1989;987:69–74. doi: 10.1016/0005-2736(89)90456-2. [DOI] [PubMed] [Google Scholar]
- 199.Lee DK, George SR, O’Dowd BF. Unravelling the roles of the apelin system: prospective therapeutic applications in heart failure and obesity. Trends Pharmacol Sci. 2006;27:190–4. doi: 10.1016/j.tips.2006.02.006. [DOI] [PubMed] [Google Scholar]
- 200.Xu N, Wang H, Fan L, Chen Q. Supraspinal administration of apelin-13 induces antinociception via the opioid receptor in mice. Peptides. 2009;30:1153–7. doi: 10.1016/j.peptides.2009.02.011. [DOI] [PubMed] [Google Scholar]
- 201.Carriba P, Ortiz O, Patkar K, Justinova Z, Stroik J, Themann A, et al. Striatal adenosine A(2A) and Cannabinoid CB1 receptors form functional heteromeric complexes that mediate the motor effects of Cannabinoids. Neuropsychopharmacology. 2007;32:2249–59. doi: 10.1038/sj.npp.1301375. [DOI] [PubMed] [Google Scholar]
- 202.Hudson BD, Hebert TE, Kelly MEM. Physical and functional interaction between CB1 cannabinoid receptors and beta(2)-adrenoceptors. Br J Pharmacol. 2010;160:627–42. doi: 10.1111/j.1476-5381.2010.00681.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Giuffrida A, Parsons LH, Kerr TM, Rodriguez de Fonseca F, Navarro M, Piomelli D. Dopamine activation of endogenous cannabinoid signaling in dorsal striatum. Nat Neurosci. 1999;2:358–63. doi: 10.1038/7268. [DOI] [PubMed] [Google Scholar]
- 204.Glass M, Felder CC. Concurrent stimulation of cannabinoid CB1 and dopamine D2 receptors augments cAMP accumulation in striatal neurons: evidence for a Gs linkage to the CB1 receptor. J Neurosci. 1997;17:5327–33. doi: 10.1523/JNEUROSCI.17-14-05327.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Jarrahian A, Watts VJ, Barker EL. D2 dopamine receptors modulate Galpha-subunit coupling of the CB1 cannabinoid receptor. J Pharmacol Exp Ther. 2004;308:880–6. doi: 10.1124/jpet.103.057620. [DOI] [PubMed] [Google Scholar]
- 206.Kearn CS, Blake-Palmer K, Daniel E, Mackie K, Glass M. Concurrent stimulation of cannabinoid CB1 and dopamine D2 receptors enhances heterodimer formation: a mechanism for receptor cross-talk? Mol Pharmacol. 2005;67:1697–704. doi: 10.1124/mol.104.006882. [DOI] [PubMed] [Google Scholar]
- 207.Ward RJ, Pediani JD, Milligan G. Heteromultimerization of cannabinoid CB1 receptor and orexin OX1 receptor generates a unique complex in which both protomers are regulated by orexin A. J Biol Chem. 2011;286:37414–28. doi: 10.1074/jbc.M111.287649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Hilairet S, Bouaboula M, Carriere D, Le Fur G, Casellas P. Hypersensitization of the orexin 1 receptor by the CB1 receptor—evidence for cross-talk blocked by the specific CB1 antagonist, SR141716. J Biol Chem. 2003;278:23731–7. doi: 10.1074/jbc.M212369200. [DOI] [PubMed] [Google Scholar]
- 209.Carriba P, Navarro G, Ciruela F, Ferre S, Casado V, Agnati L, et al. Detection of heteromerization of more than two proteins by sequential BRET-FRET. Nat Methods. 2008;5:727–33. doi: 10.1038/nmeth.1229. [DOI] [PubMed] [Google Scholar]
- 210.Martire A, Tebano MT, Chiodi V, Ferreira SG, Cunha RA, Kofalvi A, et al. Presynaptic adenosine A2A receptors control cannabinoid CB1 receptor-mediated inhibition of striatal glutamatergic neurotransmission. J Neurochem. 2011;116:273–80. doi: 10.1111/j.1471-4159.2010.07101.x. [DOI] [PubMed] [Google Scholar]
- 211.Soria G, Castane A, Berrendero F, Ledent C, Parmentier M, Maldonado R, et al. Adenosine A2A receptors are involved in physical dependence and place conditioning induced by THC. Eur J Neurosci. 2004;20:2203–13. doi: 10.1111/j.1460-9568.2004.03682.x. [DOI] [PubMed] [Google Scholar]
- 212.Yao LN, McFarland K, Fan PP, Jiang Z, Ueda T, Diamond I. Adenosine A2a blockade prevents synergy between mu-opiate and cannabinoid CB1 receptors and eliminates heroin-seeking behavior in addicted rats. Proc Natl Acad Sci USA. 2006;103:7877–82. doi: 10.1073/pnas.0602661103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Lerner TN, Horne EA, Stella N, Kreitzer AC. Endocannabinoid signaling mediates psychomotor activation by adenosine A(2A) antagonists. J Neurosci. 2010;30:2160–4. doi: 10.1523/JNEUROSCI.5844-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Justinova Z, Ferre S, Redhi GH, Mascia P, Stroik J, Quarta D, et al. Reinforcing and neurochemical effects of cannabinoid CB1 receptor agonists, but not cocaine, are altered by an adenosine A2A receptor antagonist. Addict Biol. 2011;16:405–15. doi: 10.1111/j.1369-1600.2010.00258.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Lanthier N, Horsmans Y, Leclercq IA. The metabolic syndrome: how it may influence hepatic stellate cell activation and hepatic fibrosis. Curr Opin Clin Nutr Metab Care. 2009;12:404–11. doi: 10.1097/MCO.0b013e32832c7819. [DOI] [PubMed] [Google Scholar]
- 216.Mallat A, Lotersztajn S. Endocannabinoids and liver disease. I. Endocannabinoids and their receptors in the liver. Am J Physiol Gastrointest Liver Physiol. 2008;294:G9–G12. doi: 10.1152/ajpgi.00467.2007. [DOI] [PubMed] [Google Scholar]
- 217.Siegmund SV, Schwabe RF. Endocannabinoids and liver disease. II. Endocannabinoids in the pathogenesis and treatment of liver fibrosis. Am J Physiol Gastrointest Liver Physiol. 2008;294:G357–G362. doi: 10.1152/ajpgi.00456.2007. [DOI] [PubMed] [Google Scholar]
- 218.Teixeira-Clerc F, Julien B, Grenard P, Tran Van Nhieu J, Deveaux V, Li L, et al. CB1 cannabinoid receptor antagonism: a new strategy for the treatment of liver fibrosis. Nat Med. 2006;12:671–6. doi: 10.1038/nm1421. [DOI] [PubMed] [Google Scholar]
- 219.Hillion J, Canals M, Torvinen M, Casado V, Scott R, Terasmaa A, et al. Coaggregation, cointernalization, and codesensitization of adenosine A2A receptors and dopamine D2 receptors. J Biol Chem. 2002;277:18091–7. doi: 10.1074/jbc.M107731200. [DOI] [PubMed] [Google Scholar]
- 220.Lee SP, So CH, Rashid AJ, Varghese G, Cheng R, Lanca AJ, et al. Dopamine D1 and D2 receptor co-activation generates a novel phospholipase C-mediated calcium signal. J Biol Chem. 2004;279:35671–8. doi: 10.1074/jbc.M401923200. [DOI] [PubMed] [Google Scholar]
- 221.Rashid AJ, So CH, Kong MM, Furtak T, El-Ghundi M, Cheng R, et al. D1–D2 dopamine receptor heterooligomers with unique pharmacology are coupled to rapid activation of Gq/11 in the striatum. Proc Natl Acad Sci USA. 2007;104:654–9. doi: 10.1073/pnas.0604049104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Hasbi A, Fan T, Alijaniaram M, Nguyen T, Perreault ML, O’Dowd BF, et al. Calcium signaling cascade links dopamine D1–D2 receptor heteromer to striatal BDNF production and neuronal growth. Proc Natl Acad Sci USA. 2009;106:21377–82. doi: 10.1073/pnas.0903676106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Verma V, Hasbi A, O’Dowd BF, George SR. Dopamine D1–D2 receptor heteromer-mediated calcium release is desensitized by D1 receptor occupancy with or without signal activation dual functional regulation by G protein-coupled receptor kinase 2. J Biol Chem. 2010;285:35092–103. doi: 10.1074/jbc.M109.088625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Perreault ML, Hasbi A, Alijaniaram M, Fan T, Varghese G, Fletcher PJ, et al. The dopamine D1–D2 receptor heteromer localizes in dynorphin/enkephalin neurons increased high affinity state following amphetamine and in schizophrenia. J Biol Chem. 2010;285:36625–34. doi: 10.1074/jbc.M110.159954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Fiorentini C, Busi C, Gorruso E, Gotti C, Spano P, Missale C. Reciprocal regulation of dopamine D1 and D3 receptor function and trafficking by heterodimerization. Mol Pharmacol. 2008;74:59–69. doi: 10.1124/mol.107.043885. [DOI] [PubMed] [Google Scholar]
- 226.Gines S, Hillion J, Torvinen M, Le Crom S, Casado V, Canela EI, et al. Dopamine D1 and adenosine A1 receptors form functionally interacting heteromeric complexes. Proc Natl Acad Sci USA. 2000;97:8606–11. doi: 10.1073/pnas.150241097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Scarselli M, Novi F, Schallmach E, Lin R, Baragli A, Colzi A, et al. D2/D3 dopamine receptor heterodimers exhibit unique functional properties. J Biol Chem. 2001;276:30308–14. doi: 10.1074/jbc.M102297200. [DOI] [PubMed] [Google Scholar]
- 228.Maggio R, Millan MJ. Dopamine D2–D3 receptor heteromers: pharmacological properties and therapeutic significance. Curr Opin Pharmacol. 2010;10:100–7. doi: 10.1016/j.coph.2009.10.001. [DOI] [PubMed] [Google Scholar]
- 229.Trincavelli ML, Daniele S, Orlandini E, Navarro G, Casado V, Giacomelli C, et al. A new D-2 dopamine receptor agonist allosterically modulates A(2A) adenosine receptor signalling by interacting with the A(2A)/D-2 receptor heteromer. Cell Signal. 2012;24:951–60. doi: 10.1016/j.cellsig.2011.12.018. [DOI] [PubMed] [Google Scholar]
- 230.Canals M, Marcellino D, Fanelli F, Ciruela F, de Benedetti P, Goldberg SR, et al. Adenosine A(2A)-dopamine D2 receptor-receptor heteromerization—qualitative and quantitative assessment by fluorescence and bioluminescence energy transfer. J Biol Chem. 2003;278:46741–9. doi: 10.1074/jbc.M306451200. [DOI] [PubMed] [Google Scholar]
- 231.Kamiya T, Saitoh O, Yoshioka K, Nakata H. Oligomerization of adenosine A(2A) and dopamine D-2 receptors in living cells. Biochem Biophys Res Commun. 2003;306:544–9. doi: 10.1016/s0006-291x(03)00991-4. [DOI] [PubMed] [Google Scholar]
- 232.Fuxe K, Canals M, Torvinen M, Marcellino D, Terasmaa A, Genedani S, et al. Intramembrane receptor-receptor interactions: a novel principle in molecular medicine. J Neural Transm. 2007;114:49–75. doi: 10.1007/s00702-006-0589-0. [DOI] [PubMed] [Google Scholar]
- 233.Kern A, Albarran-Zeckler R, Walsh HE, Smith RG. Apo-ghrelin receptor forms heteromers with DRD2 in hypothalamic neurons and is essential for anorexigenic effects of DRD2 agonism. Neuron. 2012;73:317–32. doi: 10.1016/j.neuron.2011.10.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Koschatzky S, Tschammer N, Gmeiner P. Cross-receptor interactions between dopamine D(2L) and neurotensin NTS(1) receptors modulate binding affinities of dopaminergics. ACS Chem Neurosci. 2011;2:308–16. doi: 10.1021/cn200020y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Thibault D, Albert PR, Pineyro G, Trudeau LE. Neurotensin triggers dopamine D2 receptor desensitization through a protein kinase C and beta-arrestin1-dependent mechanism. J Biol Chem. 2011;286:9174–84. doi: 10.1074/jbc.M110.166454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Romero-Fernandez W, Borroto-Escuela DO, Agnati LF, Fuxe K. Evidence for the existence of dopamine d2-oxytocin receptor heteromers in the ventral and dorsal striatum with facilitatory receptor-receptor interactions. Mol Psychiatry. 2012 doi: 10.1038/mp.2012.103. http://dx.doi.org/10.1038/mp.2012.103. [DOI] [PubMed]
- 237.Baragli A, Alturaihi H, Watt HL, Abdallah A, Kumar U. Heterooligomerization of human dopamine receptor 2 and somatostatin receptor 2 Co-immunoprecipitation and fluorescence resonance energy transfer analysis. Cell Signal. 2007;19:2304–16. doi: 10.1016/j.cellsig.2007.07.007. [DOI] [PubMed] [Google Scholar]
- 238.Albizu L, Holloway T, Gonzalez-Maeso J, Sealfon SC. Functional crosstalk and heteromerization of serotonin 5-HT2A and dopamine D2 receptors. Neuropharmacology. 2011;61:770–7. doi: 10.1016/j.neuropharm.2011.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Gonzalez S, Moreno-Delgado D, Moreno E, Perez-Capote K, Franco R, Mallol J, et al. Circadian-related heteromerization of adrenergic and dopamine D(4) receptors modulates melatonin synthesis and release in the pineal gland. PLoS Biol. 2012;10:e1001347. doi: 10.1371/journal.pbio.1001347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Rebois RV, Maki K, Meeks JA, Fishman PH, Hebert TE, Northup JK. D(2)-like dopamine and beta-adrenergic receptors form a signaling complex that integrates G(s)- and G(i)-mediated regulation of adenylyl cyclase. Cell Signal. 2012;24:2051–60. doi: 10.1016/j.cellsig.2012.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Dreher JK, Jackson DM. Role of D1 and D2 dopamine receptors in mediating locomotor activity elicited from the nucleus accumbens of rats. Brain Res. 1989;487:267–77. doi: 10.1016/0006-8993(89)90831-7. [DOI] [PubMed] [Google Scholar]
- 242.Kita K, Shiratani T, Takenouchi K, Fukuzako H, Takigawa M. Effects of D1 and D2 dopamine receptor antagonists on cocaine-induced self-stimulation and locomotor activity in rats. Eur Neuropsychopharmacol. 1999;9:1–7. doi: 10.1016/s0924-977x(97)00098-9. [DOI] [PubMed] [Google Scholar]
- 243.Piomelli D, Pilon C, Giros B, Sokoloff P, Martres MP, Schwartz JC. Dopamine activation of the arachidonic acid cascade as a basis for D1/D2 receptor synergism. Nature. 1991;353:164–7. doi: 10.1038/353164a0. [DOI] [PubMed] [Google Scholar]
- 244.Levavi-Sivan B, Park BH, Fuchs S, Fishburn CS. Human D3 dopamine receptor in the medulloblastoma TE671 cell line: cross-talk between D1 and D3 receptors. FEBS Lett. 1998;439:138–42. doi: 10.1016/s0014-5793(98)01356-8. [DOI] [PubMed] [Google Scholar]
- 245.Bordet R, Ridray S, Schwartz JC, Sokoloff P. Involvement of the direct striatonigral pathway in levodopa-induced sensitization in 6-hydroxydopamine-lesioned rats. Eur J Neurosci. 2000;12:2117–23. doi: 10.1046/j.1460-9568.2000.00089.x. [DOI] [PubMed] [Google Scholar]
- 246.Ridray S, Griffon N, Mignon V, Souil E, Carboni S, Diaz J, et al. Coexpression of dopamine D1 and D3 receptors in islands of Calleja and shell of nucleus accumbens of the rat: opposite and synergistic functional interactions. Eur J Neurosci. 1998;10:1676–86. doi: 10.1046/j.1460-9568.1998.00173.x. [DOI] [PubMed] [Google Scholar]
- 247.Schwartz JC, Diaz J, Bordet R, Griffon N, Perachon S, Pilon C, et al. Functional implications of multiple dopamine receptor subtypes: the D1/D3 receptor coexistence. Brain Res Brain Res Rev. 1998;26:236–42. doi: 10.1016/s0165-0173(97)00046-5. [DOI] [PubMed] [Google Scholar]
- 248.Jung MY, Schmauss C. Decreased c-fos responses to dopamine D(1) receptor agonist stimulation in mice deficient for D(3) receptors. J Biol Chem. 1999;274:29406–12. doi: 10.1074/jbc.274.41.29406. [DOI] [PubMed] [Google Scholar]
- 249.Karasinska JM, George SR, Cheng R, O’Dowd BF. Deletion of dopamine D1 and D3 receptors differentially affects spontaneous behaviour and cocaine-induced locomotor activity, reward and CREB phosphorylation. Eur J Neurosci. 2005;22:1741–50. doi: 10.1111/j.1460-9568.2005.04353.x. [DOI] [PubMed] [Google Scholar]
- 250.Ferre S, Popoli P, Gimenezllort L, Finnman UB, Martinez E, Decarolis AS, et al. Postsynaptic antagonistic interaction between adenosine a(1) and dopamine D-1 receptors. Neuroreport. 1994;6:73–6. doi: 10.1097/00001756-199412300-00020. [DOI] [PubMed] [Google Scholar]
- 251.Popoli P, Gimenez-Llort L, Pezzola A, Reggio R, Martinez E, Fuxe K, et al. Adenosine A1 receptor blockade selectively potentiates the motor effects induced by dopamine D1 receptor stimulation in rodents. Neurosci Lett. 1996;218:209–13. doi: 10.1016/s0304-3940(96)13143-8. [DOI] [PubMed] [Google Scholar]
- 252.Ferre S, Popoli P, Tinner-Staines B, Fuxe K. Adenosine A1 receptor-dopamine D1 receptor interaction in the rat limbic system: modulation of dopamine D1 receptor antagonist binding sites. Neurosci Lett. 1996;208:109–12. doi: 10.1016/0304-3940(96)12577-5. [DOI] [PubMed] [Google Scholar]
- 253.Ferre S, Torvinen M, Antoniou K, Irenius E, Civelli O, Arenas E, et al. Adenosine A1 receptor-mediated modulation of dopamine D1 receptors in stably cotransfected fibroblast cells. J Biol Chem. 1998;273:4718–24. doi: 10.1074/jbc.273.8.4718. [DOI] [PubMed] [Google Scholar]
- 254.Fink JS, Weaver DR, Rivkees SA, Peterfreund RA, Pollack AE, Adler EM, et al. Molecular cloning of the rat A2 adenosine receptor: selective co-expression with D2 dopamine receptors in rat striatum. Brain Res Mol Brain Res. 1992;14:186–95. doi: 10.1016/0169-328x(92)90173-9. [DOI] [PubMed] [Google Scholar]
- 255.Ferre S, von Euler G, Johansson B, Fredholm BB, Fuxe K. Stimulation of high-affinity adenosine A2 receptors decreases the affinity of dopamine D2 receptors in rat striatal membranes. Proc Natl Acad Sci USA. 1991;88:7238–41. doi: 10.1073/pnas.88.16.7238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Ferre S, Fuxe K. Dopamine denervation leads to an increase in the intramembrane interaction between adenosine-A2 and dopamine-D2 receptors in the neostriatum. Brain Res. 1992;594:124–30. doi: 10.1016/0006-8993(92)91036-e. [DOI] [PubMed] [Google Scholar]
- 257.Aoyama S, Kase H, Borrelli E. Rescue of locomotor impairment in dopamine D2 receptor-deficient mice by an adenosine A2A receptor antagonist. J Neurosci. 2000;20:5848–52. doi: 10.1523/JNEUROSCI.20-15-05848.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Grondin R, Bedard PJ, Tahar AH, Gregoire L, Mori A, Kase H. Antiparkinsonian effect of a new selective adenosine A(2A) receptor antagonist in MPTP-treated monkeys. Neurology. 1999;52:1673–7. doi: 10.1212/wnl.52.8.1673. [DOI] [PubMed] [Google Scholar]
- 259.Hauber W, Neuscheler P, Nagel J, Muller CE. Catalepsy induced by a blockade of dopamine D-1 or D-2 receptors was reversed by a concomitant blockade of adenosine A(2A) receptors in the caudate-putamen of rats. Eur J Neurosci. 2001;14:1287–93. doi: 10.1046/j.0953-816x.2001.01759.x. [DOI] [PubMed] [Google Scholar]
- 260.Torvinen M, Torri C, Tombesi A, Marcellino D, Watson S, Lluis C, et al. Trafficking of adenosine A2A and dopamine D2 receptors. J Mol Neurosci. 2005;25:191–200. doi: 10.1385/JMN:25:2:191. [DOI] [PubMed] [Google Scholar]
- 261.Soriano A, Ventura R, Molero A, Hoen R, Casado V, Cortes A, et al. Adenosine A2A receptor-antagonist/dopamine D2 receptor-agonist bivalent ligands as pharmacological tools to detect A2A-D2 receptor heteromers. J Med Chem. 2009;52:5590–602. doi: 10.1021/jm900298c. [DOI] [PubMed] [Google Scholar]
- 262.Jomphe C, Lemelin PL, Okano H, Kobayashi K, Trudeau LE. Bidirectional regulation of dopamine D2 and neurotensin NTS1 receptors in dopamine neurons. Eur J Neurosci. 2006;24:2789–800. doi: 10.1111/j.1460-9568.2006.05151.x. [DOI] [PubMed] [Google Scholar]
- 263.Young LJ, Lim MM, Gingrich B, Insel TR. Cellular mechanisms of social attachment. Horm Behav. 2001;40:133–8. doi: 10.1006/hbeh.2001.1691. [DOI] [PubMed] [Google Scholar]
- 264.Young LJ, Wang Z. The neurobiology of pair bonding. Nat Neurosci. 2004;7:1048–54. doi: 10.1038/nn1327. [DOI] [PubMed] [Google Scholar]
- 265.Lee HJ, Macbeth AH, Pagani JH, Young WS., 3rd Oxytocin: the great facilitator of life. Prog Neurobiol. 2009;88:127–51. doi: 10.1016/j.pneurobio.2009.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Petersson M, Diaz-Cabiale Z, Angel Narvaez J, Fuxe K, Uvnas-Moberg K. Oxytocin increases the density of high affinity alpha(2)-adrenoceptors within the hypothalamus, the amygdala and the nucleus of the solitary tract in ovariectomized rats. Brain Res. 2005;1049:234–9. doi: 10.1016/j.brainres.2005.05.034. [DOI] [PubMed] [Google Scholar]
- 267.Neumann ID. Brain oxytocin: a key regulator of emotional and social behaviours in both females and males. J Neuroendocrinol. 2008;20:858–65. doi: 10.1111/j.1365-2826.2008.01726.x. [DOI] [PubMed] [Google Scholar]
- 268.Aragona BJ, Liu Y, Curtis JT, Stephan FK, Wang Z. A critical role for nucleus accumbens dopamine in partner-preference formation in male prairie voles. J Neurosci. 2003;23:3483–90. doi: 10.1523/JNEUROSCI.23-08-03483.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Gingrich B, Liu Y, Cascio C, Wang Z, Insel TR. Dopamine D2 receptors in the nucleus accumbens are important for social attachment in female prairie voles (Microtus ochrogaster) Behav Neurosci. 2000;114:173–83. doi: 10.1037//0735-7044.114.1.173. [DOI] [PubMed] [Google Scholar]
- 270.Martin-Iverson MT, Radke JM, Vincent SR. The effects of cysteamine on dopamine-mediated behaviors: evidence for dopamine-somatostatin interactions in the striatum. Pharmacol Biochem Behav. 1986;24:1707–14. doi: 10.1016/0091-3057(86)90509-5. [DOI] [PubMed] [Google Scholar]
- 271.Rodriguez-Sanchez MN, Puebla L, Lopez-Sanudo S, Rodriguez-Martin E, Martin-Espinosa A, Rodriguez-Pena MS, et al. Dopamine enhances somatostatin receptor-mediated inhibition of adenylate cyclase in rat striatum and hippocampus. J Neurosci Res. 1997;48:238–48. doi: 10.1002/(sici)1097-4547(19970501)48:3<238::aid-jnr6>3.0.co;2-g. [DOI] [PubMed] [Google Scholar]
- 272.Izquierdo-Claros RM, Boyano-Adanez MC, Larsson C, Gustavsson L, Arilla E. Acute effects of D1- and D2-receptor agonist and antagonist drugs on somatostatin binding, inhibition of adenylyl cyclase activity and accumulation of inositol 1,4,5-trisphosphate in the rat striatum. Brain Res Mol Brain Res. 1997;47:99–107. doi: 10.1016/s0169-328x(97)00063-6. [DOI] [PubMed] [Google Scholar]
- 273.Leblanc R, Gauthier S, Gauvin M, Quirion R, Palmour R, Masson H. Neuro-behavioral effects of intrathecal somatostatinergic treatment in subhuman primates. Neurology. 1988;38:1887–90. doi: 10.1212/wnl.38.12.1887. [DOI] [PubMed] [Google Scholar]
- 274.Costa E. The role of serotonin in neurobiology. Integr Neurobiol Schizophrenia. 1960;2:175–227. doi: 10.1016/s0074-7742(08)60123-3. [DOI] [PubMed] [Google Scholar]
- 275.Nichols DE. Hallucinogens. Pharmacol Ther. 2004;101:131–81. doi: 10.1016/j.pharmthera.2003.11.002. [DOI] [PubMed] [Google Scholar]
- 276.Kim JS, Bailey MJ, Weller JL, Sugden D, Rath MF, Moller M, et al. Thyroid hormone and adrenergic signaling interact to control pineal expression of the dopamine receptor D4 gene (Drd4) Mol Cell Endocrinol. 2010;314:128–35. doi: 10.1016/j.mce.2009.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Montezinho LP, Castro MM, Duarte CB, Penschuck S, Geraldes CF, Mork A. The interaction between dopamine D2-like and beta-adrenergic receptors in the prefrontal cortex is altered by mood-stabilizing agents. J Neurochem. 2006;96:1336–48. doi: 10.1111/j.1471-4159.2005.03654.x. [DOI] [PubMed] [Google Scholar]
- 278.Ferre S, Agnati LF, Ciruela F, Lluis C, Woods AS, Fuxe K, et al. Neurotransmitter receptor heteromers and their integrative role in ‘local modules’: the striatal spine module. Brain Res Rev. 2007;55:55–67. doi: 10.1016/j.brainresrev.2007.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Ferre S, Ciruela F, Woods AS, Lluis C, Franco R. Functional relevance of neurotransmitter receptor heteromers in the central nervous system. Trends Neurosci. 2007;30:440–6. doi: 10.1016/j.tins.2007.07.001. [DOI] [PubMed] [Google Scholar]
- 280.Paquet M, Smith Y. Group I metabotropic glutamate receptors in the monkey striatum: subsynaptic association with glutamatergic and dopaminergic afferents. J Neurosci. 2003;23:7659–69. doi: 10.1523/JNEUROSCI.23-20-07659.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Popoli P, Pezzola A, Torvinen M, Reggio R, Pintor A, Scarchilli L, et al. The selective mGlu(5) receptor agonist CHPG inhibits quinpirole-induced turning in 6-hydroxydopamine-lesioned rats and modulates the binding characteristics of dopamine D(2) receptors in the rat striatum: interactions with adenosine A(2a) receptors. Neuropsychopharmacology. 2001;25:505–13. doi: 10.1016/S0893-133X(01)00256-1. [DOI] [PubMed] [Google Scholar]
- 282.Satake H, Sakai T. Recent advances and perceptions in studies of heterodimerization between G protein-coupled receptors. Protein Pept Lett. 2008;15:300–8. doi: 10.2174/092986608783744207. [DOI] [PubMed] [Google Scholar]
- 283.Bohn LM, Lefkowitz RJ, Gainetdinov RR, Peppel K, Caron MG, Lin FT. Enhanced morphine analgesia in mice lacking beta-arrestin 2. Science. 1999;286:2495–8. doi: 10.1126/science.286.5449.2495. [DOI] [PubMed] [Google Scholar]
- 284.Daniels DJ, Lenard NR, Etienne CL, Law PY, Roerig SC, Portoghese PS. Opioid-induced tolerance and dependence in mice is modulated by the distance between pharmacophores in a bivalent ligand series. Proc Natl Acad Sci USA. 2005;102:19208–13. doi: 10.1073/pnas.0506627102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285.Lenard NR, Daniels DJ, Portoghese PS, Roerig SC. Absence of conditioned place preference or reinstatement with bivalent ligands containing mu-opioid receptor agonist and delta-opioid receptor antagonist pharmacophores. Eur J Pharmacol. 2007;566:75–82. doi: 10.1016/j.ejphar.2007.02.040. [DOI] [PubMed] [Google Scholar]
- 286.Berg KA, Rowan MP, Gupta A, Sanchez TA, Silva M, Gomes I, et al. Allosteric interactions between delta and kappa opioid receptors in peripheral sensory neurons. Mol Pharmacol. 2012;81:264–72. doi: 10.1124/mol.111.072702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287.Pei L, Li S, Wang M, Diwan M, Anisman H, Fletcher PJ, et al. Uncoupling the dopamine D1–D2 receptor complex exerts antidepressant-like effects. Nat Med. 2010;16:1393–5. doi: 10.1038/nm.2263. [DOI] [PubMed] [Google Scholar]
- 288.Dziedzicka-Wasylewska M, Faron-Gorecka A, Gorecki A, Kusemider M. Mechanism of action of clozapine in the context of dopamine D1–D2 receptor hetero-dimerization—a working hypothesis. Pharmacol Rep. 2008;60:581–7. [PubMed] [Google Scholar]
- 289.Faron-Gorecka A, Gorecki A, Kusmider M, Wasylewski Z, Dziedzicka-Wasylewska M. The role of D1–D2 receptor hetero-dimerization in the mechanism of action of clozapine. Eur Neuropsychopharmacol. 2008;18:682–91. doi: 10.1016/j.euroneuro.2008.05.001. [DOI] [PubMed] [Google Scholar]
- 290.Perreault ML, O’Dowd BF, George SR. Dopamine receptor homooligomers and heterooligomers in schizophrenia. CNS Neurosci Ther. 2011;17:52–7. doi: 10.1111/j.1755-5949.2010.00228.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291.Guillin O, Abi-Dargham A, Laruelle M. Neurobiology of dopamine in schizophrenia. Integr Neurobiol Schizophrenia. 2007;78:1–39. doi: 10.1016/S0074-7742(06)78001-1. [DOI] [PubMed] [Google Scholar]
- 292.Dean B, Sundram S, Bradbury R, Scarr E, Copolov D. Studies on [H-3]CP-55940 binding in the human central nervous system: regional specific changes in density of cannabinoid-1 receptors associated with schizophrenia and cannabis use. Neuroscience. 2001;103:9–15. doi: 10.1016/s0306-4522(00)00552-2. [DOI] [PubMed] [Google Scholar]
- 293.Deckert J, Brenner M, Durany N, Zochling R, Paulus W, Ransmayr G, et al. Upregulation of striatal adenosine A(2A) receptors in schizophrenia. Neuroreport. 2003;14:313–6. doi: 10.1097/00001756-200303030-00003. [DOI] [PubMed] [Google Scholar]
- 294.Sundram S, Copolov D, Dean B. Clozapine decreases [3H] CP 55940 binding to the cannabinoid 1 receptor in the rat nucleus accumbens. Naunyn Schmiedebergs Arch Pharmacol. 2005;371:428–33. doi: 10.1007/s00210-005-1074-2. [DOI] [PubMed] [Google Scholar]
- 295.Fuxe K, Marcellino D, Borroto-Escuela DO, Guescini M, Fernandez-Duenas V, Tanganelli S, et al. Adenosine-dopamine interactions in the pathophysiology and treatment of CNS disorders. CNS Neurosci Ther. 2010;16:e18–e42. doi: 10.1111/j.1755-5949.2009.00126.x. [DOI] [PMC free article] [PubMed] [Google Scholar]